Thromb Haemost 2020; 120(01): 121-131
DOI: 10.1055/s-0039-1700518
New Technologies, Diagnostic Tools and Drugs
Georg Thieme Verlag KG Stuttgart · New York

PBMCs-Derived microRNA Signature as a Prethrombotic Status Discriminator in Stable Coronary Artery Disease

Jie Gao
1   Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
2   China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
,
Jia Liu
1   Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
2   China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
,
Ying Zhang
1   Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
2   China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
,
BaoYi Guan
1   Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
2   China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
,
Hua Qu
1   Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
2   China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
,
Hua Chai
1   Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
2   China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
,
WenTing Wang
3   Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
,
XiaoJuan Ma
1   Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
2   China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
,
DaZhuo Shi
1   Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
2   China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
› Author Affiliations
Funding This work was supported by the National Natural Science Foundation of China, grant no. 81573818 and 81774141 (to D.S.).
Further Information

Publication History

25 March 2019

03 September 2019

Publication Date:
12 November 2019 (online)

Abstract

Prethrombotic status (PTS) in patients with stable coronary artery disease (SCAD) increases the risk of coronary thrombosis. Accumulating evidences have indicated that micro-ribonucleic acids (miRNAs) may serve as promising biomarkers for SCAD patients with PTS. The present study aimed to identify the miRNA signature in SCAD patients with PTS and evaluated their diagnostic significance. In the screening phase, 32 differently expressed miRNAs (DEMs) in peripheral blood mononuclear cells (PBMCs) were detected in 35 SCAD patients compared with 5 healthy controls by microarray. MiRNA-gene network analysis was then performed, and 4 DEMs were selected for validation with reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR) test in an independent cohort comprising 79 SCAD patients and 19 healthy controls. Compared with healthy controls, RT-qPCR test verified the upregulations of miR-34a-5p, miR-432–5p, and miR-370–3p detected by microarray; while the upregulation of miR-495–3p measured by RT-qPCR was not consistent with its low expression detected by microarray. Only miR-34a-5p and miR-495–3p were significantly upregulated in the PTS group compared with the non-PTS group (p < 0.01, p < 0.05). Receiver-operating characteristic (ROC) analysis showed that PBMCs-derived miR-34a-5p and miR-495–3p may discriminate PTS with the areas under the ROC curve (AUC) of 0.780 (confidence interval [CI]95% = 0.673–0.866) and 0.712 (CI95% = 0.599–0.808), respectively. The combination of miR-34a-5p and fibrinogen (FIB, a traditional biomarker for PTS) improved AUC to 0.885 (CI95% = 0.793–0.946) and showed added predictive ability compared with FIB, with an integrated discrimination improvement of 0.201 (p < 0.01). Therefore, the combination of miR-34a-5p and FIB may serve as an efficient tool for distinguishing PTS in SCAD patients.

Supplementary Material

 
  • References

  • 1 Libby P. Mechanisms of acute coronary syndromes and their implications for therapy. N Engl J Med 2013; 368 (21) 2004-2013
  • 2 Orbe J, Zudaire M, Serrano R. , et al. Increased thrombin generation after acute versus chronic coronary disease as assessed by the thrombin generation test. Thromb Haemost 2008; 99 (02) 382-387
  • 3 Smid M, Dielis AW, Winkens M. , et al. Thrombin generation in patients with a first acute myocardial infarction. J Thromb Haemost 2011; 9 (03) 450-456
  • 4 Bauer KA, Rosenberg RD. The pathophysiology of the prethrombotic state in humans: insights gained from studies using markers of hemostatic system activation. Blood 1987; 70 (02) 343-350
  • 5 Mannucci PM, Giangrande PL. Detection of the prethrombotic state due to procoagulant imbalance. Eur J Haematol 1992; 48 (02) 65-69
  • 6 López Y, Paloma MJ, Rifón J, Cuesta B, Páramo JA. Measurement of prethrombotic markers in the assessment of acquired hypercoagulable states. Thromb Res 1999; 93 (02) 71-78
  • 7 Zheng H, Ma HP, Chen L, Zhan HT, Guo H. Prethrombotic state and cardiac events in patients with coronary heart disease during noncardiac surgery. Clin Appl Thromb Hemost 2014; 20 (01) 84-90
  • 8 Musunuru K, Hickey KT, Al-Khatib SM. , et al; American Heart Association Council on Functional Genomics and Translational Biology, Council on Clinical Cardiology, Council on Cardiovascular Disease in the Young, Council on Cardiovascular and Stroke Nursing, Council on Epidemiology and Prevention, Council on Hypertension, Council on Lifestyle and Cardiometabolic Health, Council on Quality of Care and Outcomes Research, and Stroke Council. Basic concepts and potential applications of genetics and genomics for cardiovascular and stroke clinicians: a scientific statement from the American Heart Association. Circ Cardiovasc Genet 2015; 8 (01) 216-242
  • 9 Wang J, Pei Y, Zhong Y, Jiang S, Shao J, Gong J. Altered serum microRNAs as novel diagnostic biomarkers for atypical coronary artery disease. PLoS One 2014; 9 (09) e107012
  • 10 Han H, Qu G, Han C. , et al. MiR-34a, miR-21 and miR-23a as potential biomarkers for coronary artery disease: a pilot microarray study and confirmation in a 32 patient cohort. Exp Mol Med 2015; 47: e138
  • 11 Gao J, Ma X, Zhang Y, Guo M, Shi D. The role of microRNAs in prethrombotic status associated with coronary artery disease. Thromb Haemost 2017; 117 (03) 429-436
  • 12 Montalescot G, Sechtem U, Achenbach S. , et al; Task Force Members; ESC Committee for Practice Guidelines; Document Reviewers. 2013 ESC guidelines on the management of stable coronary artery disease: the Task Force on the management of stable coronary artery disease of the European Society of Cardiology. Eur Heart J 2013; 34 (38) 2949-3003
  • 13 Zeller T, Keller T, Ojeda F. , et al. Assessment of microRNAs in patients with unstable angina pectoris. Eur Heart J 2014; 35 (31) 2106-2114
  • 14 Jakob P, Kacprowski T, Briand-Schumacher S. , et al. Profiling and validation of circulating microRNAs for cardiovascular events in patients presenting with ST-segment elevation myocardial infarction. Eur Heart J 2017; 38 (07) 511-515
  • 15 Yu S, Yue SW, Liu Z, Zhang T, Xiang N, Fu H. Yerba mate (Ilex paraguariensis) improves microcirculation of volunteers with high blood viscosity: a randomized, double-blind, placebo-controlled trial. Exp Gerontol 2015; 62: 14-22
  • 16 Sethupathy P, Megraw M, Hatzigeorgiou AG. A guide through present computational approaches for the identification of mammalian microRNA targets. Nat Methods 2006; 3 (11) 881-886
  • 17 Diez D, Wheelock AM, Goto S. , et al. The use of network analyses for elucidating mechanisms in cardiovascular disease. Mol Biosyst 2010; 6 (02) 289-304
  • 18 Corney DC, Flesken-Nikitin A, Godwin AK, Wang W, Nikitin AY. MicroRNA-34b and MicroRNA-34c are targets of p53 and cooperate in control of cell proliferation and adhesion-independent growth. Cancer Res 2007; 67 (18) 8433-8438
  • 19 Pencina MJ, D'Agostino Sr RB, D'Agostino Jr RB, Vasan RS. Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond. Stat Med 2008; 27 (02) 157-172 , discussion 207–212
  • 20 Stokes KY, Granger DN. Platelets: a critical link between inflammation and microvascular dysfunction. J Physiol 2012; 590 (05) 1023-1034
  • 21 Sueta D, Hokimoto S, Ogawa H. Could perioperative bleeding complications be predicted using a platelet aggregation test?. Thromb Res 2015; 136 (03) 491-492
  • 22 Breet NJ, van Werkum JW, Bouman HJ. , et al. Comparison of platelet function tests in predicting clinical outcome in patients undergoing coronary stent implantation. JAMA 2010; 303 (08) 754-762
  • 23 Peace AJ, Tedesco AF, Foley DP, Dicker P, Berndt MC, Kenny D. Dual antiplatelet therapy unmasks distinct platelet reactivity in patients with coronary artery disease. J Thromb Haemost 2008; 6 (12) 2027-2034
  • 24 Rosenberg RD. The biochemistry and pathophysiology of the prethrombotic state. Annu Rev Med 1987; 38: 493-508
  • 25 Brainin P, Frestad D, Prescott E. The prognostic value of coronary endothelial and microvascular dysfunction in subjects with normal or non-obstructive coronary artery disease: a systematic review and meta-analysis. Int J Cardiol 2018; 254: 1-9
  • 26 Taqueti VR, Di Carli MF. Coronary microvascular disease pathogenic mechanisms and therapeutic options: JACC State-of-the-Art Review. J Am Coll Cardiol 2018; 72 (21) 2625-2641
  • 27 Antonios TF, Kaski JC, Hasan KM, Brown SJ, Singer DR. Rarefaction of skin capillaries in patients with anginal chest pain and normal coronary arteriograms. Eur Heart J 2001; 22 (13) 1144-1148
  • 28 Yuksel S, Pancar Yuksel E, Yenercag M. , et al. Abnormal nail fold capillaroscopic findings in patients with coronary slow flow phenomenon. Int J Clin Exp Med 2014; 7 (04) 1052-1058
  • 29 Taqueti VR, Hachamovitch R, Murthy VL. , et al. Global coronary flow reserve is associated with adverse cardiovascular events independently of luminal angiographic severity and modifies the effect of early revascularization. Circulation 2015; 131 (01) 19-27
  • 30 Zanatta E, Famoso G, Boscain F. , et al. Nailfold avascular score and coronary microvascular dysfunction in systemic sclerosis: a newsworthy association. Autoimmun Rev 2019; 18 (02) 177-183
  • 31 Landry P, Plante I, Ouellet DL, Perron MP, Rousseau G, Provost P. Existence of a microRNA pathway in anucleate platelets. Nat Struct Mol Biol 2009; 16 (09) 961-966
  • 32 McManus DD, Freedman JE. MicroRNAs in platelet function and cardiovascular disease. Nat Rev Cardiol 2015; 12 (12) 711-717
  • 33 Pu M, Chen J, Tao Z. , et al. Regulatory network of miRNA on its target: coordination between transcriptional and post-transcriptional regulation of gene expression. Cell Mol Life Sci 2019; 76 (03) 441-451
  • 34 Xu Y, Zhu W, Sun Y, Wang Z, Yuan W, Du Z. Functional network analysis reveals versatile microRNAs in human heart. Cell Physiol Biochem 2015; 36 (04) 1628-1643
  • 35 Canales RD, Luo Y, Willey JC. , et al. Evaluation of DNA microarray results with quantitative gene expression platforms. Nat Biotechnol 2006; 24 (09) 1115-1122
  • 36 Gao W, He HW, Wang ZM. , et al. Plasma levels of lipometabolism-related miR-122 and miR-370 are increased in patients with hyperlipidemia and associated with coronary artery disease. Lipids Health Dis 2012; 11: 55
  • 37 Zhou T, Xiang DK, Li SN, Yang LH, Gao LF, Feng C. MicroRNA-495 ameliorates cardiac microvascular endothelial cell injury and inflammatory reaction by suppressing the NLRP3 inflammasome signaling pathway. Cell Physiol Biochem 2018; 49 (02) 798-815
  • 38 Liu D, Zhang XL, Yan CH. , et al. MicroRNA-495 regulates the proliferation and apoptosis of human umbilical vein endothelial cells by targeting chemokine CCL2. Thromb Res 2015; 135 (01) 146-154
  • 39 Welten SMJ, de Jong RCM, Wezel A. , et al. Inhibition of 14q32 microRNA miR-495 reduces lesion formation, intimal hyperplasia and plasma cholesterol levels in experimental restenosis. Atherosclerosis 2017; 261: 26-36
  • 40 Liang J, Huang W, Cai W. , et al. Inhibition of microRNA-495 enhances therapeutic angiogenesis of human induced pluripotent stem cells. Stem Cells 2017; 35 (02) 337-350
  • 41 Nagalla S, Shaw C, Kong X. , et al. Platelet microRNA-mRNA coexpression profiles correlate with platelet reactivity. Blood 2011; 117 (19) 5189-5197
  • 42 Osman A, Fälker K. Characterization of human platelet microRNA by quantitative PCR coupled with an annotation network for predicted target genes. Platelets 2011; 22 (06) 433-441
  • 43 Su G, Sun G, Liu H, Shu L, Liang Z. Downregulation of miR-34a promotes endothelial cell growth and suppresses apoptosis in atherosclerosis by regulating Bcl-2. Heart Vessels 2018; 33 (10) 1185-1194
  • 44 Li Q, Kim YR, Vikram A. , et al. P66Shc-induced microRNA-34a causes diabetic endothelial dysfunction by downregulating Sirtuin1. Arterioscler Thromb Vasc Biol 2016; 36 (12) 2394-2403
  • 45 Fan W, Fang R, Wu X. , et al. Shear-sensitive microRNA-34a modulates flow-dependent regulation of endothelial inflammation. J Cell Sci 2015; 128 (01) 70-80
  • 46 Hart M, Walch-Rückheim B, Friedmann KS. , et al. miR-34a: a new player in the regulation of T cell function by modulation of NF-κB signaling. Cell Death Dis 2019; 10 (02) 46
  • 47 Salloum-Asfar S, Arroyo AB, Teruel-Montoya R. , et al. MiRNA-based regulation of hemostatic factors through hepatic nuclear factor-4 alpha. PLoS One 2016; 11 (05) e0154751
  • 48 Navarro F, Gutman D, Meire E. , et al. miR-34a contributes to megakaryocytic differentiation of K562 cells independently of p53. Blood 2009; 114 (10) 2181-2192
  • 49 Nakanishi K, Fukuda S, Tanaka A. , et al. Persistent epicardial adipose tissue accumulation is associated with coronary plaque vulnerability and future acute coronary syndrome in non-obese subjects with coronary artery disease. Atherosclerosis 2014; 237 (01) 353-360
  • 50 Tousoulis D, Papageorgiou N, Androulakis E, Briasoulis A, Antoniades C, Stefanadis C. Fibrinogen and cardiovascular disease: genetics and biomarkers. Blood Rev 2011; 25 (06) 239-245
  • 51 Ellins EA, Rees DA, Deanfield JE, Steptoe A, Halcox JP. Increased fibrinogen responses to psychophysiological stress predict future endothelial dysfunction implications for cardiovascular disease?. Brain Behav Immun 2017; 60: 233-239
  • 52 Wannamethee SG, Whincup PH, Lennon L, Papacosta O, Lowe GD. Associations between fibrin D-dimer, markers of inflammation, incident self-reported mobility limitation, and all-cause mortality in older men. J Am Geriatr Soc 2014; 62 (12) 2357-2362