Circulating microRNAs, novel biomarkers of acute myocardial infarction: a system

时间:2022-10-03 08:23:07

BACKGROUND: MicroRNAs (MiRNA) are a novel class of non-coding RNAs involved in the regulation of gene expression post-transcriptionally by cleavage or translational repression of their specific target miRNAs. Numerous studies have demonstrated that circulating miRNAs are stable and abundant in blood and aberrantly expressed under pathological conditions, including cardiovascular diseases. The implications of circulating miRNAs in acute myocardial infarction have recently been recognized. This review will highlight the potential role of miRNA as a novel class of biomarkers in acute myocardial infarction.

METHODS: This systemic review is based on our own work and other related reports.

RESULTS: During diseases circulating miRNAs are derived from not only circulating blood cells but also other tissues affected by ongoing diseases. These disease-related miRNAs in the blood can serve as potential biomarkers.

CONCLUSION: The circulating miRNAs can be used as novel biomarkers potentially offering more sensitive and specific tests than those currently available for diagnosis of acute myocardial infarction.

KEY WORDS: MicroRNAs; Acute myocardial infarction; Biomarkers

World J Emerg Med 2012;3(4):257–260

DOI: 10.5847/ wjem.j.1920-8642.2012.04.003

INTRODUCTION

A rapid diagnosis of acute myocardial infarction (AMI) is critical for appropriate management of patients with chest pain. Clinical presentation of echocardiography findings and cardiac markers are often nonspecific in patients with chest pain.[1] Therefore the ideal biomarker for rapid and reliable diagnosis of AMI is still lacking. Human serum and other body fluids are rich resources for the identification of novel biomarkers, which can be measured in routine clinical diagnosis. Over many decades, it has been shown that cell-free DNA and RNA are present in serum and other body fluids and that these circulating nucleic acids may represent potential biomarkers.[2,3] MicroRNAs (MiRNAs) are a novel class of non-coding RNAs that are widely expressed in plants and animals to regulate gene expression post-transcriptionally by cleavage or translational repression of their specific target miRNAs.[4] The expression of miRNAs is often tissue-specific or developmental specific and thus miRNAs play an important role in repression of gene expression at specific stages in various biological processes.

In the past years, we have witnessed the rapid development of many creative-innovative-inventive techniques and methodologies pertinent to miRNA research and applications in cardiovascular disease.[5,6] These technologies have convincingly demonstrated their efficacy and reliability in being novel biomarkers of cardiovascular disease. Here we summarize the rapidly expanding knowledge of the circulating miRNAs in acute myocardial infarction.

miRNA biogenesis and function

The human genome has been estimated to encode up to 1000 miRNAs that are predicted to regulate a third of human genes. According to the current understanding, miRNAs originate from a primary transcript (hundreds to thousands of nucleotides long) called a pri-miRNA which is transcribed by RNA polymerase II and regulated by transcription factors as same as conventional mRNAs; after nuclear cleavage by Drosha (a ribonuclease III)the pri-miRNA binds protein DGCR8/Pasha to generate a hairpin-shaped pre-miRNA;[7,8] the nuclear export factor exportin-5 transports the pre-miRNA to the cytoplasm;[9,10] and within the cytoplasm the ribonuclease III Dicer and its cofactors (PACT and TRBP) process the precursors into 19- to 25-nucleotide miRNA duplexes. The double-stranded RNA molecule dissociates and one strand is incorporated into the RNA-induced silencing complex (RISC). The miRNA-loaded RISC is capable of bringing the target mRNAs to inhibit their translation by cleavage and degradation of mRNA or by blocking translation through several different mechanisms.[11]

Stabilization and possible release mechanisms

While miRNA presence is relevant for the regulation of several genes in tissues the possibility to extract and reliably determine cell-free miRNA content in body fluids like serum as was shown in many studies.[12,13] The findings of the studies confirmed that miRNAs are enriched in the small RNA fraction isolated from serum samples. Because of the high levels of RNase activity in plasma synthetic miRNAs were quickly degraded,[14] but cell-free miRNAs was stable under harsh conditions including boiling low/high pH extended storage and multiple freeze-thaw cycles.[11–14] Filtering and differential centrifugation experiments suggest that miRNAs are not derived from cells circulating in the blood.[15] At present there are at least two possible explanations for the origin of circulating miRNAs:

One hypothesis is that miRNAs are released during tissue injury. For example, miRNA-208, which exclusively expressed in the heart, was measured in the serum after heart tissue injury.[16] Alternatively circulating miRNAs might have biological functions, e.g. acting as long-distance signals as is known from plants.[17]

MiRNAs are detected in serum or plasma in a remarkably stable form and are resistant against RNase-mediated degradation.[14,18] Increasing evidence suggests that there are several different mechanisms that protect miRNAs from degradation. One possible mechanism is that the miRNAs are incorporated in lipid vesicles to avoid degradation. Several types of small lipid vesicles released by cells are described: microvesicles/microparticles (relatively large 100 nm to 1 ?m) are shed from the cell plasma membrane into the extracellular space and released into the blood stream under physiological and pathological conditions. The exosomes (smaller membrane fragments 30–100 nm) are derived from the endosomal compartment. In addition, apoptotic bodies (larger up to 4 ?m) are released when cells are undergoing apoptotic death.[19]

Functional roles of circulating miRNAs

Increasing studies suggest that circulating miRNAs can regulate target gene expression. Cells can take up extracellular RNAas shown for RNA which had been incorporated into microvesicles. In physiological and pathological conditions secreted microvesicles containing miRNAs can transfer the miRNAs to recipient cells and regulate target gene expression.[20]

Darnell et al[21] confirmed that the expression of miR-1 coincided with the onset of cardiomyocyte differentiation in the developing heart tube and somitic myotome development in chick embryos. In a study[22] exosomes were identified as an active component of conditioned medium and cardiomyocyte progenitor cells released exosomes to stimulate the migration of endothelial cells. The authors of the study also indicated that endothelial cell-specific miR-126 plays an essential role in neoangiogenesis after MI. Moreover, it was shown that glioblastoma-derived microvesicles enriched with mRNAs and miRNAs can be taken up by brain microvascular endothelial cells with subsequent alteration of the genetic endothelial program.[23]

Circulating miRNAs are stable and abundant in blood. It is believed that circulating extracellular miRNAs play important roles in cell-cell communication. However, the functional roles of circulating miRNAs and proposed mechanisms remain to be determined.

Circulating miRNAs reflecting physiological and pathological changes

With direct cloning and microarray-based profiling, a large set of miRNAs expressed in the heart have been identified in vivo and ex vitro. MiR-1 and miR-133 were found to be highly enriched in cardiac and skeletal muscle. Increasingly, miR-1 and miR-133 involved in cardiac hypertrophy were identified.[24] da Costa Martins et al[25] demonstrated that miR-199b is a direct calcineurin/NFAT target gene which increases in expression in mouse and human heart failure.

Multiple miRNAs aberrantly expressed in infarcted hearts are responsible for cardiac remodeling after MI or I/R injury. Ren et al[26] determined the miRNA signature in ischemic hearts by a mouse model of cardiac I/R in vivo and ex vivo, and they found that miR-320 expression was consistently dysregulated after ischemia. Further studies from the same group showed that the miR-144/451 cluster also protected against simulated I/R-induced cardiomyocyte death.[27] It was found that some miRNAs such as miR-29miR-199 and miRNA-24 were downregulated after myocardial infarction.[28–30]

MiRNAs also regulate the expression of molecules involved in regulating action potentials and cardiac conduction. In a study,[31] miR-1 was found to be upregulated in the patients with coronary artery disease (CAD) and in rat ischemic hearts and confirmed its involvement in arrhythmogenesis. Recently, Lu et al[32] reported that miR-328 contributes to adverse electrical remodeling in atrial fibrillation (AF). These studies strongly highlight the potential of miRNAs as a novel biomarker.

Circulating miRNAs as biomarkers for acute myocardial infarction

Muscle-specific and cardiac-specific miRNAs in plasma of AMI patients were significantly higher than those of healthy subjects cornary heart disease (CHD) patients without AMI or patients with other cardiovascular diseases. miRNA-208b and miR-499 are expressed by myosin heavy chains in cardiac or skeletal muscle and both are used for detecting cardiac damage.[33,34] In a study,[35] miRNA-208b and miR-499 were highly increased in MI patients (>105-fold, P< 0.001) and they were detected in healthy controls. One hour early after the onset of chest pain, the two miRNAs were detected and their diagnostic accuracy is robust. In addition, in patients who presented less than 3 hours after the onset of chest pain, miR-499 was positive in 93% of the patients and hs-cTnT in 88% (P=0.78). Patients with ST-elevation MI (n=397) had a higher miRNA concentration than those with non-ST-elevation MI (n=113) (P

Recent studies revealed that other miRNAs which were not muscle specific or cardiac specific have been overlooked. MiR-328 is ubiquitously distributed in many tissues and has been found to involve in many pathological conditions. Wang et al[36] found that the miR-133 and miR-328 levels in plasma from AMI patients exhibited respectively a 4.4-fold and 10.9-fold increase compared with those from healthy controls. The elevated circulating miR-133 and miR-328 levels were recovered to the control levels at 7 days after AMI. ROC analysis revealed that the AUCs of miR-328 in plasma and whole blood were 0.810 and 0.872, suggesting that the increased miR-328 level might be associated with AMI. In other studies, miR-1miR-1291miR-663bmiR-145 and miR-30c levels were increased in both experimental AMI models and/or in patients with AMI.[37–38]

In conclusion, the blood contains large amounts of stable miRNAs derived from various tissues/organs and circulating miRNAs are resistant to RNaseA digestion and other harsh conditions. However, the mechanism of resistance of miRNAs to RNase requires further study. Studies unequivocally showed that circulating miRNAs can be used as a novel biomarker potentially offering more sensitive and specific tests than those currently available for diagnosis of acute myocardial infarction.

Since the results of studies have clearly indicated the expression of the miRNAs in blood from patients with AMI, their physiological functions and relationship with genesis need further investigation. The results also strongly suggest that during diseases circulating miRNAs are derived from not only circulating blood cells but also other tissues affected by ongoing diseases and that these disease-related miRNAs in the blood can serve as potential biomarkers. These novel biomarkers have the potential to revolutionize the present clinical management. Given the fact that miRNAs are identified as the first class of RNAs stably present in the blood, it would be of great interest to understand the biological functions of circulating miRNAs and their other application.

Funding: This work was supported grants from the National Natural Science Foundation of China (81000076) and Priority Academic Program Development of Jiangsu Higher Education Institutions (JX10231081).

Ethical approval: Not needed.

Conflicts of interest: There is no conflict of interest in this study.

Contributors: Chen Y proposed the studyand wrote the first draft. All authors read and approved the final version.

REFERENCES

1 Omland T, de Lemos JA, Sabatine MS, Christophi CA, Rice MM, Jablonski KA, et al. A sensitive cardiac troponin T assay in stable coronary artery disease. N Engl J Med 2009; 361: 2538–2547.

2 Swarup V, Rajeswari MR. Circulating (cell-free) nucleic acids–a promisingnon-invasive tool for early detection of several human diseases. FEBS Lett 2007; 581: 795–799.

3 Tsang JC, Lo YM. Circulating nucleic acids in plasma/serum. Pathology 2007; 39: 197–207.

4 Bartel DP. MicroRNAs: genomics biogenesis mechanism and function. Cell 2004; 116: 281–297.

5 van Rooij E, Marshall WS, Olson EN. Toward microRNA-based therapeutics for heart disease: the sense in antisense. Circ Res 2008; 103: 919–928.

6 Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, et al. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Research 2008; 18: 997–1006.

7 Lee Y, Jeon K, Lee JT, Kim S, Kim VN. MicroRNA maturation: stepwise processing and subcellular localization. Embo J 2002; 21: 4663–4670.

8 Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003; 425: 415–419.

9 Kim VN. MicroRNA precursors in motion: exportin-5 mediates their nuclear export. Trends Cell Biol 2004; 14: 156–159.

10 Zeng Y, Cullen BR. Structural requirements for pre-microRNA binding and nuclear export by Exportin 5. Nucleic Acids Res 2004; 32: 4776–4785.

11 Berezikov E, Guryev V, van de Belt J, Wienholds E, Plasterk RH, Cuppen E, et al. Phylogenetic shadowing and computational identification of human microRNA genes. Cell 2005; 120: 21–24.

12 Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Liggins AP, Pulford K, et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br J Haematol 2008; 141: 672–675.

13 Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, et al. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 2008; 18: 997–1006.

14 Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA 2008; 105: 10513–10518.

15 Gilad S, Meiri E, Yogev Y, Benjamin S, Lebanony D, Yerushalmi N, et al. Serum microRNAs are promising novel biomarkers. PLoS ONE 2008; 3: e3148.

16 Ji X, Takahashi R, Hiura Y, Hirokawa G, Fukushima Y, Iwai N, et al. Plasma miR-208 as a biomarker of myocardial injury. Clin Chem 2009; 55: 1944–1949.

17 Hyun TK, Uddin MN, Rim Y, Kim JY. Cell-to-cell trafficking of RNA and RNA silencing through plasmodesmata. Protoplasma 2011; 248: 101–116.

18 Gilad S, Meiri E, Yogev Y, Benjamin S, Lebanony D, Yerushalmi N, et al. Serum microRNAs are promising novel biomarkers. PLoS ONE 2008; 3: e3148.

19 Mause SF, Weber C. Microparticles: protagonists of a novel communication network for intercellular information exchange. Circ Res 2010; 107: 1047–1057.

20 Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007; 9: 654–659.

21 Darnell DK, Kaur S, Stanislaw S. MicroRNA expression during chick embryo development. Dev Dyn 2006; 235: 3156–3165.

22 Vrijsen KR, Sluijter JP, Schuchardt MW, van Balkom BW, Noort WA, Chamuleau SA, et al. Cardiomyocyte progenitor cell-derived exosomes stimulate migration of endothelial cells. J Cell Mol Med 2010; 14: 1064–1070.

23 Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol 2008; 10: 1470–1476.

24 Feng Y, Yu X. Cardinal roles of miRNA in cardiac development and disease. Sci China Life Sci 2011; 54: 1113–1120.

25 Da CMP, Salic K, Gladka MM. MicroRNA-199b targets the nuclear kinase Dyrk1a in an auto-amplification loop promoting calcineurin/NFAT signalling. Nat Cell Biol 2010; 12: 1220–1227.

26 Ren XP, Wu J, Wang X. MicroRNA-320 is involved in the regulation of cardiac ischemia/reperfusion injury by targeting heat-shock protein 20. Circulation 2009; 119: 2357–2366.

27 Zhang X, Wang X, Zhu H. Synergistic effects of the GATA-4-mediated miR-144/451 cluster in protection against simulated ischemia/reperfusion-induced cardiomyocyte death. J Mol Cell Cardiol 2010; 49: 841–850.

28 Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation 1986; 74: 1124–1136.

29 Rane S, He M, Sayed D, Vashistha H, Malhotra A, Sadoshima J, et al. Downregulation of miR-199a derepresses hypoxia-inducible factor-1alpha and Sirtuin 1 and recapitulates hypoxia preconditioning in cardiac myocytes. Circ Res 2009; 104: 879–886.

30 Qian L, Van Laake LW, Huang Y. miR-24 inhibits apoptosis and represses Bim in mouse cardiomyocytes. J Exp Med 2011; 208: 549–560.

31 Yang B, Lin H, Xiao J. The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat Med 2007; 13: 486–491.

32 Lu Y, Zhang Y, Wang N, Pan Z, Gao X, Zhang F, et al. MicroRNA-328 contributes to adverse electrical remodeling in atrial fibrillation. Circulation 2010; 122: 2378–2387.

33 van Rooij E, Quiat D, Johnson BA, Sutherland LB, Qi X, Richardson JA, et al. A family of microRNAs encoded by myosin genes governs myosin expression and muscle performance. Dev Cell 2009; 17: 662–673.

34 Fichtlscherer S, De Rosa S, Fox HS, Shwietz T, Fischer A, Liebetrau C, et al. Circulating micro-RNAs in patients with coronary artery disease. Circ Res 2010; 107: 677–684.

35 Devaux Y, Vausort M, Goretti E, Nazarov PV, Azuaje F, Gilson G, et al. Use of Circulating MicroRNAs to Diagnose Acute Myocardial Infarction. Clin Chem 2012; 58: 559–567. Epub 2012 Jan 17.

36 Wang R, Li N, Zhang Y, Ran Y, Pu J. Circulating MicroRNAs are promising novel biomarkers of acute myocardial infarction. Intern Med 2011; 50: 1789–1795.

37 Kuwabara Y, Ono K, Horie T, Nishi H, Nagao K, Kinoshita M, et al. Increased microRNA-1 and microRNA-133a levels in serum of patients with cardiovascular disease indicate the existence of myocardial damage. Circ Cardiovasc Genet 2011; 4: 446–454.

38 Meder B, Keller A, Vogel B, Haas J, Sedaghat-Hamedani F, Kayvanpour E, et al. MicroRNA signatures in total peripheral blood as novel biomarkers for acute myocardial infarction. Basic Res Cardiol 2011; 106: 13–23.

Received April 3, 2012

Accepted after revision September 20, 2012

上一篇:Do mannequin chests provide an accurate rep... 下一篇:新航线的开辟者

文档上传者
热门推荐 更多>
精品范文更多>