Journal List > Korean J Physiol Pharmacol > v.18(5) > 1025964

Li, Dong, Wang, and Wu: MicroRNA-1 in Cardiac Diseases and Cancers

Abstract

MicroRNAs (miRs) are endogenous ≈22-nt non-coding RNAs that participate in the regulation of gene expression at post-transcriptional level. MiR-1 is one of the muscle-specific miRs, aberrant expression of miR-1 plays important roles in many physiological and pathological processes. In this review, we focus on the recent studies about miR-1 in cardiac diseases and cancers. The findings indicate that miR-1 may be a novel, important biomarker, and a potential therapeutic target in cardiac diseases and cancers.

ABBREVIATIONS

miR

microRNA

UTR

untranslated region

TAGLN2

transgelin 2

MI

myocardial infarction

IR

ischemia-reperfusion

miR-1-ES

transplanting miR-1 transfected embryonic stem cells

STEMI

ST elevation myocardial infarction

RasGAP

Ras GTPase-activating protein

Cdk9

cyclin-dependent kinase 9

PNP

purine nucleoside phosphorylase

XPO6

exportin-6

INTRODUCTION

MicroRNAs (miRs) are a group of endogenous ≈22-nt non-coding RNAs that participate in the regulation of gene expression through binding to the 3'-untranslated regions (UTRs) of target mRNA at post-transcriptional level [1]. MiRs were first discovered in the nematode Caenorhabditis elegans by Rosalind C. Lee in 1993 [2]. Since then, abundant miRs were found in many species such as plants, animals and human being [3,4,5], and regarded as a class of highly conserved RNA in the course of evolution. So far, more than 1000 miRs are predicted in human genome, and regulate about 30% of human genes [6,7]. There is growing evidence that miRs play important roles in various physiological and pathological processes, including cardiogenesis, neural differentiation, cell apoptosis and tumor formation, etc [8,9,10]. Among the known miRs, miR-1 is one of the muscle-specific miRs, and highly expressed in cardiac tissue [11].
MiR-1 is encoded by two nearly identical genes: miR-1-1 and miR-1-2, which are located within intron 2 of an uncharacterized gene C20ORF166 and intron 12 of the E3 ubiquitin-protein ligase MIB1 (Mindbomb Homogog 1) on chromosomes 20 and 18, respectively [12,13]. Both miR-1-1 and miR-1-2 give rise to identical mature miR-1 species and appear to target the same mRNAs [14,15,16]. Targeted deletion of miR-1-1 results in a phenotype similar to that described for miR-1-2-null mice [14]. Accordingly, mice that lack of the miR-1-2 gene will lead to various heart abnormalities, but generally survive because they still generate some miR-1 from their remaining miR-1-1 gene [12]. When lacking both miR-1-1 and miR-1-2, mice will appear more profound abnormalities and die before weaning [8,12].
It has been reported that over-expression of miR-1 in the developing heart resulted in a decreased pool of proliferating ventricular cardiomyocytes through reducing the protein levels of Hand2, a transcription factor that promotes ventricular cardiomyocyte expansion [15]. In miR-1-2-null mice, the hearts have a series of abnormalities, including ventricular septal defect, cardiac rhythm disturbances and a striking myocyte cell-cycle abnormality [8]. These findings suggest that the proper expression of miR-1 is necessary for cardiogenesis and maintenance of the normal heart function. Changes in miR-1 expression would lead to the occurrence of many cardiac diseases.
In addition to the pathophysiological roles of miR-1 in cardiac diseases, many studies have also demonstrated that aberrant expression of miR-1 plays important roles in the initiation, development and metastasis of human cancers [17]. A large number of studies have shown that miR-1 expression is frequently down-regulated in several cancer types, which results in the up-regulation of multiple oncogenes including Slug, MET and transgelin 2 (TAGLN2), etc [18]. Restoration of miR-1 expression or transfecting miR-1 was able to significantly inhibit tumor cell proliferation, migration and invasion [18,19]. The over-expression of miR-1 has also been reported to induce tumor cell apoptosis by activating caspases-3, a key enzyme in the apoptosis cell-signaling cascade [20]. The above investigations suggested that miR-1 function as a tumor suppressor. Therefore, increasing miR-1 expression may be an effective strategy for prevention of the initiation and development of cancers.

MiR-1 AND CARDIAC DISEASES

MiR-1 and arrhythmia

Arrhythmia is one of the major reasons for sudden cardiac death. Recently, several studies have observed that the abnormal expression of miR-1 is involved in occurrence of arrhythmia. Girmatsion et al. [21] reported that the levels of miR-1 were greatly reduced in left atria from patients with persistent atrial fibrillation. While the mRNA and protein expression of inwardly rectifying potassium channel 2.1 (a target of miR-1) was significantly increased, which led to increased inwardly rectifying potassium current. In rat ventricular myocytes, over-expression of miR-1 can inhibit the protein phosphatase PP2A regulatory subunit B56α, which could enhance the functional activity of RyR2 channels and thus result in increased cardiac excitation-contraction coupling gain, elevated diastolic sarcoplasmic reticulum Ca2+ leak, and reduced sarcoplasmic reticulum Ca2+ content and promote arrhythmogenic disturbances in myocyte Ca2+ cycling [22]. In addition, Zhao et al. [8] found that deletion of miR-1-2 resulted in severe arrhythmia through up-regulating the expression of Irx5, a member of Iroquois family of homeodomain-containing transcription factors that regulates cardiac repolarization by repressing Kcnd2 (a key potassium channel) [23].

MiR-1 and myocardial infarction

Many recent studies have shown evidence for the role of miR-1 in myocardial infarction (MI). The level of miR-1 was significantly down-regulated both in MI patients and ischemia-reperfusion (IR) rat [24,25]. Transplanting miR-1 transfected embryonic stem cells (miR-1-ES) into the border zone of the infracted heart of c7BL/6 mice, the heart function was significantly improved after 2wk post-MI. The beneficial effect of miR-1-ES is attributed to protection of host myocardium from MI-induced apoptosis through activating p-AKT and inhibiting caspase-3, phosphatase and tensin homolog, and superoxide production [26]. More recently, reports show that miR-1 also exists in the circulating blood of humans and animals [27]. Under physiological conditions, miR-1 is present at very low levels in plasma. However, the level of miR-1 is significantly increased both in MI patients and mice [6]. Using a rat model of AMI, Cheng et al. [28] found that the serum miR-1 was rapidly increased after AMI with a peak at 6 h, and returned to basal levels at 3 day, there was a strong positive correlation between serum miR-1 level and myocardial infarct size. Circulating miR-1 in ST elevation myocardial infarction (STEMI) patients was increased 300-fold compared with healthy controls [29]. It is worth mentioning that increased circulating miR-1 was not related to age, gender, blood pressure, diabetes mellitus or the established biomarkers for AMI. Circulating miR-1, therefore, may be a novel, independent biomarker for early diagnosis of AMI [30].

MiR-1 and cardiac hypertrophy

Cardiac hypertrophy is a pathological condition and characterized by increased cell size, enhanced protein synthesis, and heightened organization of the sarcomere [31]. It is a persistent response of heart to a variety of harmful stimulation, including increased sympathetic nerve activity and pressure load [32,33]. Recent studies indicated that aberrant expression of miR-1 plays an important role in the development of cardiac hypertrophy. Both in rat hypertrophic left ventricle and phenylephrine-induced hypertrophic cardiomyocytes, miR-1 level was significantly decreased concomitantly with an increased expression of twinfilin-1(a cytoskeleton regulatory protein was identified as a potential target of miR-1), which stimulates hypertrophy through regulation of cardiac cytoskeleton. Further investigation found that silencing of endogenous miR-1 could induce cardiomyocyte hypertrophy. Whereas over-expression of miR-1 could reduce cell size and decrease the expression of hypertrophy-associated genes such as Acta1, Myh7 and Nppa, calmodulin, Mef2a, Ras GTPase-activating protein (RasGAP) and cyclin-dependent kinase 9 (Cdk9), etc [34,35,36]. These findings indicate that miR-1 exerts an anti-hypertrophic property.

MiR-1 and heart failure

Matured cardiomyocytes are the terminally differentiated cells, lacking of reproductive activity. The excessive apoptosis of cardiomyocytes would lead to the decreasing of cardiomyocytes, which is extensively recognized as an important mechanism of heart failure [37]. There is growing evidence that cardiomyocyte apoptosis is closely related to the abnormal expression of miR-1. It has been reported that treatment of cultured cardiomyocytes with H2O2 could significantly increase the rate of apoptotic cells concomItantly with up-regulated expression of miR-1. Further experiments found that over-expression of miR-1 aggravated H2O2-induced cardiomyocyte apoptosis, while inhibition of miR-1 using antisense inhibitory oligonucleotides resulted in significant resistance to H2O2 [38]. The miR-1 expression level was also markedly increased in the model of high glucose-induced cardiomyocyte apoptosis [39].

MiR-1 AND CANCERS

MiR-1 and rhabdomyosarcoma

Rhabdomyosarcomas are the most common soft tissue sarcomas in children [40]. Since the muscle-specific nature of miR-1, many studies focused on the roles of miR-1 in the development of rhabdomyosarcomas. Yan et al. [41] demonstrated that the miR-1 expression was significantly decreased in rhabdomyosarcomas compared with normal skeletal muscle, and presented at very low levels in a rhabdomyosarcoma cell line. At the same time, the expression of c-Met, a target of miR-1, was markedly up-regulated. There was a significant inverse correlation between miR-1 and c-Met expression. c-Met is a disulfide-linked heterodimer and encoded by MET oncogene [42,43]. The increased expression and activity of c-MET exist in different tumors, and contribute to tumor growth, invasiveness and metastasis [44]. In over-expression of miR-1 rhabdomyosarcoma cells, the cell growth and migration was significantly inhibited, accompanied by the down-regulated expression of c-MET [41]. Decreased level of miR-1 in rhabdomyosarcoma cells was also proved by Rao et al. [40]. Meanwhile, they revealed that transcriptional profiling of cells after miR-1 expression exerts a strong promyogenic influence on poorly differentiated rhabdomyosarcoma cells.

MiR-1 and lung cancer

Lung cancer is still the number one cause of cancer-related death worldwide [45]. Since the 5-year survival rate was no more than 15%, it is necessary for early diagnosis and therapy of lung cancer. It has been reported that miR-1 was also expressed in lung, although at a lower level than in the skeletal muscle and heart [46]. There is evidence that miR-1 was significantly down-regulated in vinyl carbamate-induced mouse lung tumors [47]. Nasser et al. [20] also demonstrated that miR-1 level was markedly decreased in primary human lung cancer, including different histological types such as non-small lung cancers, adenocarcinomas, squamous cell carcinormas, large cell carcinoma and bronchoalveolar cell carcinoma. Consistent with the results in vivo, miR-1 level was almost undetectable in 16 lung cancer cell lines, which is relatively high in non-tumorigenic bronchial epithelial. Moreover, a recent study indicated that low serum expression level of miR-1 was related to unfavorable prognosis, and may be used as a biomarker to predict non-small-cell lung cancer survival [48].

MiR-1 and bladder cancer

Bladder cancer is seventh most common cancer in the world and the second most cancer of the genitourinary tract [49,50]. Although most of patients have a good clinical prognosis, about 50% of cases will suffer recurrence within four years of their initial diagnosis [51]. Therefore, elucidating the pathogenesis is helpful to prevent recurrence of bladder cancer. Recent studies found that the expression of miR-1 was significantly down-regulated in clinical bladder cancer specimens compared with normal bladder tissue [52]. In miR-1-transfected bladder cancer cell lines, the cell viability, migration and invasion were markedly inhibited, while the cell apoptosis was significantly increased [52,53]. To investigate the underlying anti-cancer mechanisms of miR-1, the authors observed the expression of TAGLN2 and LASP1, which were directly regulated by miR-1 and exert a potential oncogenic effect. The findings showed that the level of TAGLN2 and LASP1 was remarkably repressed in miR-1-transfected bladder cancer cell lines.

MiR-1 and prostate cancer

Recently, several studies have observed changes in expression of miR-1 in prostate cancer. Kojima et al. [54] reported that the expression levels of miR-1 were significantly decreased in prostate cancer compared with nonprostate cancer tissues. Restoration of miR-1 in prostate cancer cells could markedly suppress cell proliferation, migration and invasion. The beneficial effect of miR-1 was associated with regulation of purine nucleoside phosphorylase (PNP), which is identified as a new target of miR-1 and recognized as a potential oncogene. Ambs et al. [55] also demonstrated the down-regulation of miR-1 in prostate cancer, accompanied by increased exportin-6 (XPO6). This result was further verified in vitro, transfection of the prostate cancer cells with miR-1 could inhibit protein expression of XPO6.
Accumulating evidence indicate that dysregulated expression of miR-1 is a frequently event in cancers. In addition to the cancers described above, the abnormal expression of miR-1 was also found in other cancers, including colon cancer, hepatocelluar carcinoma, and head and squamous cell carcinoma, etc [56,57,58]. It is worthy to note that miR-1 expression was always down-regulated in all of these cancers. These investigations suggest that miR-1 may be a novel, potential target for tumor therapies.

CONCLUSION

MiR-1 is important in the development of heart and skeletal muscle. Dysregulated expression of miR-1 plays an important role in the pathogenesis of cardiac diseases and cancers. The underlying mechanisms are closely related to negatively regulation of its target genes, including Irx5, twinfilin-1, transgelin 2 and XPO6, etc. Although numerous studies have demonstrated the importance of miR-1 in cardiac diseases and cancers, and also recognized it as a novel, important biomarker and a potential therapeutic target, the exact role and mechanism of miR-1 is still not very clear due to the complexity of these diseases. More studies are needed to elucidate the role and mechanism of miR-1 in cardiac diseases and cancers.

ACKNOWLEDGEMENTS

This work was supported by the National Nature Science Foundation of China [81301057 to J.H.W. and 81260211 to Z.P.W.] and the Guangxi Nature Science Foundation of China [2013GXNSFBA019126 to J. Z. Li] and the Bureau of Public Health of Guangxi Province [Z2013206].

ABBREVIATIONS

miR

microRNA

UTR

untranslated region

TAGLN2

transgelin 2

MI

myocardial infarction

IR

ischemia-reperfusion

miR-1-ES

transplanting miR-1 transfected embryonic stem cells

STEMI

ST elevation myocardial infarction

RasGAP

Ras GTPase-activating protein

Cdk9

cyclin-dependent kinase 9

PNP

purine nucleoside phosphorylase

XPO6

exportin-6

References

1. Vimalraj S, Selvamurugan N. MicroRNAs expression and their regulatory networks during mesenchymal stem cells differentiation toward osteoblasts. Int J Biol Macromol. 2014; 66:194–202. PMID: 24560946.
crossref
2. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993; 75:843–854. PMID: 8252621.
crossref
3. Wu G. Plant microRNAs and development. J Genet Genomics. 2013; 40:217–230. PMID: 23706297.
crossref
4. Qian J, Zhang Z, Liang J, Ge Q, Duan X, Ma F, Li F. The full-length transcripts and promoter analysis of intergenic microRNAs in Drosophila melanogaster. Genomics. 2011; 97:294–303. PMID: 21333734.
crossref
5. Quach H, Barreiro LB, Laval G, Zidane N, Patin E, Kidd KK, Kidd JR, Bouchier C, Veuille M, Antoniewski C, Quintana-Murci L. Signatures of purifying and local positive selection in human miRNAs. Am J Hum Genet. 2009; 84:316–327. PMID: 19232555.
crossref
6. D'Alessandra Y, Devanna P, Limana F, Straino S, Di Carlo A, Brambilla PG, Rubino M, Carena MC, Spazzafumo L, De Simone M, Micheli B, Biglioli P, Achilli F, Martelli F, Maggiolini S, Marenzi G, Pompilio G, Capogrossi MC. Circulating microRNAs are new and sensitive biomarkers of myocardial infarction. Eur Heart J. 2010; 31:2765–2773. PMID: 20534597.
7. Valeri N, Vannini I, Fanini F, Calore F, Adair B, Fabbri M. Epigenetics, miRNAs, and human cancer: a new chapter in human gene regulation. Mamm Genome. 2009; 20:573–580. PMID: 19697081.
crossref
8. Zhao Y, Ransom JF, Li A, Vedantham V, von Drehle M, Muth AN, Tsuchihashi T, McManus MT, Schwartz RJ, Srivastava D. Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miRNA-1-2. Cell. 2007; 129:303–317. PMID: 17397913.
crossref
9. Montalban E, Mattugini N, Ciarapica R, Provenzano C, Savino M, Scagnoli F, Prosperini G, Carissimi C, Fulci V, Matrone C, Calissano P, Nasi S. MiR-21 is an Ngf-modulated microRNA that supports Ngf signaling and regulates neuronal degeneration in PC12 cells. Neuromolecular Med. 2014; 16:415–430. PMID: 24492999.
crossref
10. Aguda BD. Modeling microRNA-transcription factor networks in cancer. Adv Exp Med Biol. 2013; 774:149–167. PMID: 23377973.
crossref
11. Malizia AP, Wang DZ. MicroRNAs in cardiomyocyte development. Wiley Interdiscip Rev Syst Biol Med. 2011; 3:183–190. PMID: 21305703.
crossref
12. Heidersbach A, Saxby C, Carver-Moore K, Huang Y, Ang YS, de Jong PJ, Ivey KN, Srivastava D. microRNA-1 regulates sarcomere formation and suppresses smooth muscle gene expression in the mammalian heart. Elife. 2013; 2:e01323. PMID: 24252873.
crossref
13. Fu JD, Rushing SN, Lieu DK, Chan CW, Kong CW, Geng L, Wilson KD, Chiamvimonvat N, Boheler KR, Wu JC, Keller G, Hajjar RJ, Li RA. Distinct roles of microRNA-1 and -499 in ventricular specification and functional maturation of human embryonic stem cell-derived cardiomyocytes. PLoS One. 2011; 6:e27417. PMID: 22110643.
crossref
14. Tao G, Martin JF. MicroRNAs get to the heart of development. Elife. 2013; 2:e01710. PMID: 24252874.
crossref
15. Zhao Y, Samal E, Srivastava D. Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature. 2005; 436:214–220. PMID: 15951802.
crossref
16. Mishima Y, Stahlhut C, Giraldez AJ. miR-1-2 gets to the heart of the matter. Cell. 2007; 129:247–249. PMID: 17448987.
crossref
17. Nohata N, Hanazawa T, Enokida H, Seki N. microRNA-1/133a and microRNA-206/133b clusters: dysregulation and functional roles in human cancers. Oncotarget. 2012; 3:9–21. PMID: 22308266.
crossref
18. Osaka E, Yang X, Shen JK, Yang P, Feng Y, Mankin HJ, Hornicek FJ, Duan Z. MicroRNA-1 (miR-1) inhibits chordoma cell migration and invasion by targeting slug. J Orthop Res. 2014; 32:1075–1082. PMID: 24760686.
crossref
19. Yu QQ, Wu H, Huang X, Shen H, Shu YQ, Zhang B, Xiang CC, Yu SM, Guo RH, Chen L. MiR-1 targets PIK3CA and inhibits tumorigenic properties of A549 cells. Biomed Pharmacother. 2014; 68:155–161. PMID: 24486107.
crossref
20. Nasser MW, Datta J, Nuovo G, Kutay H, Motiwala T, Majumder S, Wang B, Suster S, Jacob ST, Ghoshal K. Down-regulation of micro-RNA-1 (miR-1) in lung cancer. Suppression of tumorigenic property of lung cancer cells and their sensitization to doxorubicin-induced apoptosis by miR-1. J Biol Chem. 2008; 283:33394–33405. PMID: 18818206.
crossref
21. Girmatsion Z, Biliczki P, Bonauer A, Wimmer-Greinecker G, Scherer M, Moritz A, Bukowska A, Goette A, Nattel S, Hohnloser SH, Ehrlich JR. Changes in microRNA-1 expression and IK1 up-regulation in human atrial fibrillation. Heart Rhythm. 2009; 6:1802–1809. PMID: 19959133.
crossref
22. Terentyev D, Belevych AE, Terentyeva R, Martin MM, Malana GE, Kuhn DE, Abdellatif M, Feldman DS, Elton TS, Györke S. miR-1 overexpression enhances Ca2+ release and promotes cardiac arrhythmogenesis by targeting PP2A regulatory subunit B56alpha and causing CaMKII-dependent hyperphosphorylation of RyR2. Circ Res. 2009; 104:514–521. PMID: 19131648.
23. Costantini DL, Arruda EP, Agarwal P, Kim KH, Zhu Y, Zhu W, Lebel M, Cheng CW, Park CY, Pierce SA, Guerchicoff A, Pollevick GD, Chan TY, Kabir MG, Cheng SH, Husain M, Antzelevitch C, Srivastava D, Gross GJ, Hui CC, Backx PH, Bruneau BG. The homeodomain transcription factor Irx5 establishes the mouse cardiac ventricular repolarization gradient. Cell. 2005; 123:347–358. PMID: 16239150.
crossref
24. Bostjancic E, Zidar N, Stajer D, Glavac D. MicroRNAs miR-1, miR-133a, miR-133b and miR-208 are dysregulated in human myocardial infarction. Cardiology. 2010; 115:163–169. PMID: 20029200.
crossref
25. He B, Xiao J, Ren AJ, Zhang YF, Zhang H, Chen M, Xie B, Gao XG, Wang YW. Role of miR-1 and miR-133a in myocardial ischemic postconditioning. J Biomed Sci. 2011; 18:22. PMID: 21406115.
crossref
26. Glass C, Singla DK. MicroRNA-1 transfected embryonic stem cells enhance cardiac myocyte differentiation and inhibit apoptosis by modulating the PTEN/Akt pathway in the infarcted heart. Am J Physiol Heart Circ Physiol. 2011; 301:H2038–H2049. PMID: 21856911.
crossref
27. Sayed AS, Xia K, Yang TL, Peng J. Circulating microRNAs: a potential role in diagnosis and prognosis of acute myocardial infarction. Dis Markers. 2013; 35:561–566. PMID: 24249943.
crossref
28. Cheng Y, Tan N, Yang J, Liu X, Cao X, He P, Dong X, Qin S, Zhang C. A translational study of circulating cell-free microRNA-1 in acute myocardial infarction. Clin Sci (Lond). 2010; 119:87–95. PMID: 20218970.
crossref
29. Gidlöf O, Andersson P, van der Pals J, Götberg M, Erlinge D. Cardiospecific microRNA plasma levels correlate with troponin and cardiac function in patients with ST elevation myocardial infarction, are selectively dependent on renal elimination, and can be detected in urine samples. Cardiology. 2011; 118:217–226. PMID: 21701171.
crossref
30. Ai J, Zhang R, Li Y, Pu J, Lu Y, Jiao J, Li K, Yu B, Li Z, Wang R, Wang L, Li Q, Wang N, Shan H, Li Z, Yang B. Circulating microRNA-1 as a potential novel biomarker for acute myocardial infarction. Biochem Biophys Res Commun. 2010; 391:73–77. PMID: 19896465.
crossref
31. Kee HJ, Kook H. Roles and targets of class I and IIa histone deacetylases in cardiac hypertrophy. J Biomed Biotechnol. 2011; 2011:928326. PMID: 21151616.
crossref
32. Cha HN, Choi JH, Kim YW, Kim JY, Ahn MW, Park SY. Metformin inhibits isoproterenol-induced cardiac hypertrophy in mice. Korean J Physiol Pharmacol. 2010; 14:377–384. PMID: 21311678.
crossref
33. Rifki OF, Bodemann BO, Battiprolu PK, White MA, Hill JA. RalGDS-dependent cardiomyocyte autophagy is required for load-induced ventricular hypertrophy. J Mol Cell Cardiol. 2013; 59:128–138. PMID: 23473774.
crossref
34. Li Q, Song XW, Zou J, Wang GK, Kremneva E, Li XQ, Zhu N, Sun T, Lappalainen P, Yuan WJ, Qin YW, Jing Q. Attenuation of microRNA-1 derepresses the cytoskeleton regulatory protein twinfilin-1 to provoke cardiac hypertrophy. J Cell Sci. 2010; 123:2444–2452. PMID: 20571053.
crossref
35. Ikeda S, He A, Kong SW, Lu J, Bejar R, Bodyak N, Lee KH, Ma Q, Kang PM, Golub TR, Pu WT. MicroRNA-1 negatively regulates expression of the hypertrophy-associated calmodulin and Mef2a genes. Mol Cell Biol. 2009; 29:2193–2204. PMID: 19188439.
crossref
36. Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M. MicroRNAs play an essential role in the development of cardiac hypertrophy. Circ Res. 2007; 100:416–424. PMID: 17234972.
crossref
37. Yang C, Liu Z, Liu K, Yang P. Mechanisms of Ghrelin anti-heart failure: inhibition of Ang II-induced cardiomyocyte apoptosis by down-regulating AT1R expression. PLoS One. 2014; 9:e85785. PMID: 24465706.
crossref
38. Tang Y, Zheng J, Sun Y, Wu Z, Liu Z, Huang G. MicroRNA-1 regulates cardiomyocyte apoptosis by targeting Bcl-2. Int Heart J. 2009; 50:377–387. PMID: 19506341.
crossref
39. Yu XY, Song YH, Geng YJ, Lin QX, Shan ZX, Lin SG, Li Y. Glucose induces apoptosis of cardiomyocytes via microRNA-1 and IGF-1. Biochem Biophys Res Commun. 2008; 376:548–552. PMID: 18801338.
crossref
40. El Nadi E, Moussa EA, Zekri W, Taha H, Yones A, Zaghloul MS, El Wakeel M, Labib RM. Outcome of Rhabdomyosarcoma in First Year of Life: Children's Cancer Hospital 57357 Egypt. Sarcoma. 2013; 2013:439213. PMID: 23983569.
crossref
41. Yan D, Dong Xda E, Chen X, Wang L, Lu C, Wang J, Qu J, Tu L. MicroRNA-1/206 targets c-Met and inhibits rhabdomyosarcoma development. J Biol Chem. 2009; 284:29596–29604. PMID: 19710019.
crossref
42. Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003; 4:915–925. PMID: 14685170.
crossref
43. Mazzone M, Comoglio PM. The Met pathway: master switch and drug target in cancer progression. FASEB J. 2006; 20:1611–1621. PMID: 16873884.
crossref
44. Danilkovitch-Miagkova A, Zbar B. Dysregulation of Met receptor tyrosine kinase activity in invasive tumors. J Clin Invest. 2002; 109:863–867. PMID: 11927612.
45. Kim KC, Lee C. Reversal of Cisplatin resistance by epigallocatechin gallate is mediated by downregulation of axl and tyro 3 expression in human lung cancer cells. Korean J Physiol Pharmacol. 2014; 18:61–66. PMID: 24634598.
crossref
46. Mishima T, Mizuguchi Y, Kawahigashi Y, Takizawa T, Takizawa T. RT-PCR-based analysis of microRNA (miR-1 and -124) expression in mouse CNS. Brain Res. 2007; 1131:37–43. PMID: 17182009.
crossref
47. Melkamu T, Zhang X, Tan J, Zeng Y, Kassie F. Alteration of microRNA expression in vinyl carbamate-induced mouse lung tumors and modulation by the chemopreventive agent indole-3-carbinol. Carcinogenesis. 2010; 31:252–258. PMID: 19748927.
crossref
48. Hu Z, Chen X, Zhao Y, Tian T, Jin G, Shu Y, Chen Y, Xu L, Zen K, Zhang C, Shen H. Serum microRNA signatures identified in a genome-wide serum microRNA expression profiling predict survival of non-small-cell lung cancer. J Clin Oncol. 2010; 28:1721–1726. PMID: 20194856.
crossref
49. Shimada K, Fujii T, Anai S, Fujimoto K, Konishi N. ROS generation via NOX4 and its utility in the cytological diagnosis of urothelial carcinoma of the urinary bladder. BMC Urol. 2011; 11:22. PMID: 22032647.
crossref
50. Al-Sukhun S, Hussain M. Current understanding of the biology of advanced bladder cancer. Cancer. 2003; 97(8 Suppl):2064–2075. PMID: 12673698.
crossref
51. Babjuk M, Oosterlinck W, Sylvester R, Kaasinen E, Böhle A, Palou-Redorta J. European Association of Urology (EAU). EAU guidelines on non-muscle-invasive urothelial carcinoma of the bladder. Eur Urol. 2008; 54:303–314. PMID: 18468779.
crossref
52. Chiyomaru T, Enokida H, Kawakami K, Tatarano S, Uchida Y, Kawahara K, Nishiyama K, Seki N, Nakagawa M. Functional role of LASP1 in cell viability and its regulation by microRNAs in bladder cancer. Urol Oncol. 2012; 30:434–443. PMID: 20843712.
crossref
53. Yoshino H, Chiyomaru T, Enokida H, Kawakami K, Tatarano S, Nishiyama K, Nohata N, Seki N, Nakagawa M. The tumour-suppressive function of miR-1 and miR-133a targeting TAGLN2 in bladder cancer. Br J Cancer. 2011; 104:808–818. PMID: 21304530.
crossref
54. Kojima S, Chiyomaru T, Kawakami K, Yoshino H, Enokida H, Nohata N, Fuse M, Ichikawa T, Naya Y, Nakagawa M, Seki N. Tumour suppressors miR-1 and miR-133a target the oncogenic function of purine nucleoside phosphorylase (PNP) in prostate cancer. Br J Cancer. 2012; 106:405–413. PMID: 22068816.
crossref
55. Ambs S, Prueitt RL, Yi M, Hudson RS, Howe TM, Petrocca F, Wallace TA, Liu CG, Volinia S, Calin GA, Yfantis HG, Stephens RM, Croce CM. Genomic profiling of microRNA and messenger RNA reveals deregulated microRNA expression in prostate cancer. Cancer Res. 2008; 68:6162–6170. PMID: 18676839.
crossref
56. Migliore C, Martin V, Leoni VP, Restivo A, Atzori L, Petrelli A, Isella C, Zorcolo L, Sarotto I, Casula G, Comoglio PM, Columbano A, Giordano S. MiR-1 downregulation cooperates with MACC1 in promoting MET overexpression in human colon cancer. Clin Cancer Res. 2012; 18:737–747. PMID: 22179665.
57. Turato C, Simonato D, Quarta S, Gatta A, Pontisso P. MicroRNAs and SerpinB3 in hepatocellular carcinoma. Life Sci. 2014; 100:9–17. PMID: 24496037.
crossref
58. Nohata N, Sone Y, Hanazawa T, Fuse M, Kikkawa N, Yoshino H, Chiyomaru T, Kawakami K, Enokida H, Nakagawa M, Shozu M, Okamoto Y, Seki N. miR-1 as a tumor suppressive microRNA targeting TAGLN2 in head and neck squamous cell carcinoma. Oncotarget. 2011; 2:29–42. PMID: 21378409.
crossref
TOOLS
Similar articles