From Molecular Mechanisms to Therapeutics: Understanding MicroRNA-21 in Cancer
Abstract
:1. Introduction
2. MicroRNA-21 and Its Role in Cancer
3. Regulation of MicroRNA-21 Biogenesis
4. Regulation of Stemness Markers and Differentiation by MicroRNA-21
5. MicroRNA-21 in Cell Death
6. MicroRNA-21 in Chemoresistance
7. MicroRNA-21 as a Biomarker in Cancer
8. MicroRNA-21-Targeted RNA Therapeutics
9. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
References
- Ambros, V.; Bartel, B.; Bartel, D.P.; Burge, C.B.; Carrington, J.C.; Chen, X.; Dreyfuss, G.; Eddy, S.R.; Griffiths-Jones, S.; Marshall, M.; et al. A uniform system for microRNA annotation. RNA 2003, 9, 277–279. [Google Scholar] [CrossRef]
- Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef]
- Kozomara, A.; Birgaoanu, M.; Griffiths-Jones, S. miRBase: From microRNA sequences to function. Nucleic Acids Res. 2019, 47, D155–D162. [Google Scholar] [CrossRef]
- Alles, J.; Fehlmann, T.; Fischer, U.; Backes, C.; Galata, V.; Minet, M.; Hart, M.; Abu-Halima, M.; Grässer, F.A.; Lenhof, H.P.; et al. An estimate of the total number of true human miRNAs. Nucleic Acids Res. 2019, 47, 3353–3364. [Google Scholar] [CrossRef] [PubMed]
- Friedman, R.C.; Farh, K.K.; Burge, C.B.; Bartel, D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009, 19, 92–105. [Google Scholar] [CrossRef]
- Borchert, G.M.; Lanier, W.; Davidson, B.L. RNA polymerase III transcribes human microRNAs. Nat. Struct. Mol. Biol. 2006, 13, 1097–1101. [Google Scholar] [CrossRef] [PubMed]
- Cai, X.; Hagedorn, C.H.; Cullen, B.R. Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA 2004, 10, 1957–1966. [Google Scholar] [CrossRef] [PubMed]
- Han, J.; Lee, Y.; Yeom, K.H.; Kim, Y.K.; Jin, H.; Kim, V.N. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 2004, 18, 3016–3027. [Google Scholar] [CrossRef]
- Gregory, R.I.; Yan, K.P.; Amuthan, G.; Chendrimada, T.; Doratotaj, B.; Cooch, N.; Shiekhattar, R. The Microprocessor complex mediates the genesis of microRNAs. Nature 2004, 432, 235–240. [Google Scholar] [CrossRef]
- O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yi, R.; Qin, Y.; Macara, I.G.; Cullen, B.R. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev. 2003, 17, 3011–3016. [Google Scholar] [CrossRef]
- Gregory, R.I.; Chendrimada, T.P.; Cooch, N.; Shiekhattar, R. Human RISC couples microRNA biogenesis and posttranscriptional gene silencing. Cell 2005, 123, 631–640. [Google Scholar] [CrossRef]
- Meijer, H.A.; Smith, E.M.; Bushell, M. Regulation of miRNA strand selection: Follow the leader? Biochem. Soc. Trans. 2014, 42, 1135–1140. [Google Scholar] [CrossRef]
- Lee, D.; Park, D.; Park, J.H.; Kim, J.H.; Shin, C. Poly(A)-specific ribonuclease sculpts the 3′ ends of microRNAs. RNA 2019, 25, 388–405. [Google Scholar] [CrossRef]
- Wyman, S.K.; Knouf, E.C.; Parkin, R.K.; Fritz, B.R.; Lin, D.W.; Dennis, L.M.; Krouse, M.A.; Webster, P.J.; Tewari, M. Post-transcriptional generation of miRNA variants by multiple nucleotidyl transferases contributes to miRNA transcriptome complexity. Genome Res. 2011, 21, 1450–1461. [Google Scholar] [CrossRef] [PubMed]
- Yoda, M.; Cifuentes, D.; Izumi, N.; Sakaguchi, Y.; Suzuki, T.; Giraldez, A.J.; Tomari, Y. Poly(A)-specific ribonuclease mediates 3′-end trimming of Argonaute2-cleaved precursor microRNAs. Cell Rep. 2013, 5, 715–726. [Google Scholar] [CrossRef]
- Yin, J.; Kim, T.H.; Park, N.; Shin, D.; Choi, H.I.; Cho, S.; Park, J.B.; Kim, J.H. TRIM71 suppresses tumorigenesis via modulation of Lin28B-let-7-HMGA2 signaling. Oncotarget 2016, 7, 79854–79868. [Google Scholar] [CrossRef]
- Lee, S.H.; Cho, S.; Kim, M.S.; Choi, K.; Cho, J.Y.; Gwak, H.S.; Kim, Y.J.; Yoo, H.; Lee, S.H.; Park, J.B.; et al. The ubiquitin ligase human TRIM71 regulates let-7 microRNA biogenesis via modulation of Lin28B protein. Biochim. Biophys. Acta 2014, 1839, 374–386. [Google Scholar] [CrossRef]
- Huang, J.T.; Wang, J.; Srivastava, V.; Sen, S.; Liu, S.M. MicroRNA Machinery Genes as Novel Biomarkers for Cancer. Front. Oncol. 2014, 4, 113. [Google Scholar] [CrossRef] [PubMed]
- López-Urrutia, E.; Bustamante Montes, L.P.; Ladrón de Guevara Cervantes, D.; Pérez-Plasencia, C.; Campos-Parra, A.D. Crosstalk Between Long Non-coding RNAs, Micro-RNAs and mRNAs: Deciphering Molecular Mechanisms of Master Regulators in Cancer. Front. Oncol. 2019, 9, 669. [Google Scholar] [CrossRef]
- Park, J.Y.; Lee, J.E.; Park, J.B.; Yoo, H.; Lee, S.H.; Kim, J.H. Roles of Long Non-Coding RNAs on Tumorigenesis and Glioma Development. Brain Tumor Res. Treat. 2014, 2, 1–6. [Google Scholar] [CrossRef]
- Makarova, J.A.; Shkurnikov, M.U.; Wicklein, D.; Lange, T.; Samatov, T.R.; Turchinovich, A.A.; Tonevitsky, A.G. Intracellular and extracellular microRNA: An update on localization and biological role. Prog. Histochem. Cytochem. 2016, 51, 33–49. [Google Scholar] [CrossRef] [PubMed]
- Salzman, J. Circular RNA Expression: Its Potential Regulation and Function. Trends Genet. 2016, 32, 309–316. [Google Scholar] [CrossRef]
- Meng, S.; Zhou, H.; Feng, Z.; Xu, Z.; Tang, Y.; Li, P.; Wu, M. CircRNA: Functions and properties of a novel potential biomarker for cancer. Mol. Cancer 2017, 16, 94. [Google Scholar] [CrossRef] [PubMed]
- Xin, R.; Gao, Y. isoCirc catalogs full-length circular RNA isoforms in human transcriptomes. Nat. Commun. 2021, 12, 266. [Google Scholar] [CrossRef]
- Kim, T.K.; Hemberg, M.; Gray, J.M.; Costa, A.M.; Bear, D.M.; Wu, J.; Harmin, D.A.; Laptewicz, M.; Barbara-Haley, K.; Kuersten, S.; et al. Widespread transcription at neuronal activity-regulated enhancers. Nature 2010, 465, 182–187. [Google Scholar] [CrossRef] [PubMed]
- Melo, C.A.; Drost, J.; Wijchers, P.J.; van de Werken, H.; de Wit, E.; Vrielink, J.A.O.; Elkon, R.; Melo, S.A.; Léveillé, N.; Kalluri, R.; et al. eRNAs are required for p53-dependent enhancer activity and gene transcription. Mol. Cell 2013, 49, 524–535. [Google Scholar] [CrossRef] [PubMed]
- Siomi, M.C.; Sato, K.; Pezic, D.; Aravin, A.A. PIWI-interacting small RNAs: The vanguard of genome defence. Nat. Rev. Mol. Cell Biol 2011, 12, 246–258. [Google Scholar] [CrossRef]
- Morris, K.V.; Chan, S.W.; Jacobsen, S.E.; Looney, D.J. Small interfering RNA-induced transcriptional gene silencing in human cells. Science 2004, 305, 1289–1292. [Google Scholar] [CrossRef] [PubMed]
- Matera, A.G.; Terns, R.M.; Terns, M.P. Non-coding RNAs: Lessons from the small nuclear and small nucleolar RNAs. Nat. Rev. Mol. Cell Biol. 2007, 8, 209–220. [Google Scholar] [CrossRef]
- Ender, C.; Krek, A.; Friedländer, M.R.; Beitzinger, M.; Weinmann, L.; Chen, W.; Pfeffer, S.; Rajewsky, N.; Meister, G. A human snoRNA with microRNA-like functions. Mol. Cell 2008, 32, 519–528. [Google Scholar] [CrossRef]
- Theimer, C.A.; Feigon, J. Structure and function of telomerase RNA. Curr. Opin. Struct. Biol. 2006, 16, 307–318. [Google Scholar] [CrossRef]
- Haussecker, D.; Huang, Y.; Lau, A.; Parameswaran, P.; Fire, A.Z.; Kay, M.A. Human tRNA-derived small RNAs in the global regulation of RNA silencing. RNA 2010, 16, 673–695. [Google Scholar] [CrossRef] [PubMed]
- Persson, H.; Kvist, A.; Vallon-Christersson, J.; Medstrand, P.; Borg, A.; Rovira, C. The non-coding RNA of the multidrug resistance-linked vault particle encodes multiple regulatory small RNAs. Nat. Cell Biol. 2009, 11, 1268–1271. [Google Scholar] [CrossRef]
- Gulìa, C.; Signore, F.; Gaffi, M.; Gigli, S.; Votino, R. Y RNA: An Overview of Their Role as Potential Biomarkers and Molecular Targets in Human Cancers. Cancers 2020, 12, 1238. [Google Scholar] [CrossRef] [PubMed]
- Christov, C.P.; Trivier, E.; Krude, T. Noncoding human Y RNAs are overexpressed in tumours and required for cell proliferation. Br. J. Cancer 2008, 98, 981–988. [Google Scholar] [CrossRef] [PubMed]
- Fujita, S.; Ito, T.; Mizutani, T.; Minoguchi, S.; Yamamichi, N.; Sakurai, K.; Iba, H. miR-21 Gene expression triggered by AP-1 is sustained through a double-negative feedback mechanism. J. Mol. Biol. 2008, 378, 492–504. [Google Scholar] [CrossRef]
- Feng, Y.H.; Tsao, C.J. Emerging role of microRNA-21 in cancer. Biomed. Rep. 2016, 5, 395–402. [Google Scholar] [CrossRef]
- Bailey, M.H.; Tokheim, C.; Porta-Pardo, E.; Sengupta, S.; Bertrand, D.; Weerasinghe, A.; Colaprico, A.; Wendl, M.C.; Kim, J.; Reardon, B.; et al. Comprehensive Characterization of Cancer Driver Genes and Mutations. Cell 2018, 173, 371–385. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Guo, D.; Zhang, Y. Association of MicroRNA-21 with p53 at Mutant Sites R175H and R248Q, Clinicopathological Features, and Prognosis of NSCLC. Mol. Ther. Oncolytics 2020, 19, 208–217. [Google Scholar] [CrossRef]
- Bornachea, O.; Santos, M.; Martínez-Cruz, A.B.; García-Escudero, R.; Dueñas, M.; Costa, C.; Segrelles, C.; Lorz, C.; Buitrago, A.; Saiz-Ladera, C.; et al. EMT and induction of miR-21 mediate metastasis development in Trp53-deficient tumours. Sci. Rep. 2012, 2, 434. [Google Scholar] [CrossRef] [PubMed]
- Chu, N.J.; Anders, R.A.; Fertig, E.J.; Cao, M.; Hopkins, A.C.; Keenan, B.P. Inhibition of miR-21 Regulates Mutant KRAS Effector Pathways and Intercepts Pancreatic Ductal Adenocarcinoma Development. Cancer Prev. Res. 2020, 13, 569–582. [Google Scholar] [CrossRef]
- Hatley, M.E.; Patrick, D.M.; Garcia, M.R.; Richardson, J.A.; Bassel-Duby, R.; van Rooij, E.; Olson, E.N. Modulation of K-Ras-dependent lung tumorigenesis by MicroRNA-21. Cancer Cell 2010, 18, 282–293. [Google Scholar] [CrossRef] [PubMed]
- Seike, M.; Goto, A.; Okano, T.; Bowman, E.D.; Schetter, A.J.; Horikawa, I.; Mathe, E.A.; Jen, J.; Yang, P.; Sugimura, H.; et al. MiR-21 is an EGFR-regulated anti-apoptotic factor in lung cancer in never-smokers. Proc. Natl. Acad. Sci. USA 2009, 106, 12085–12090. [Google Scholar] [CrossRef]
- Shen, J.; Xia, W.; Khotskaya, Y.B.; Huo, L.; Nakanishi, K.; Lim, S.O.; Du, Y.; Wang, Y.; Chang, W.C.; Chen, C.H.; et al. EGFR modulates microRNA maturation in response to hypoxia through phosphorylation of AGO2. Nature 2013, 497, 383–387. [Google Scholar] [CrossRef]
- Bloomston, M.; Frankel, W.L.; Petrocca, F.; Volinia, S.; Alder, H.; Hagan, J.P.; Liu, C.G.; Bhatt, D.; Taccioli, C.; Croce, C.M. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA 2007, 297, 1901–1908. [Google Scholar] [CrossRef]
- Iorio, M.V.; Ferracin, M.; Liu, C.G.; Veronese, A.; Spizzo, R.; Sabbioni, S.; Magri, E.; Pedriali, M.; Fabbri, M.; Campiglio, M.; et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005, 65, 7065–7070. [Google Scholar] [CrossRef]
- Schetter, A.J.; Leung, S.Y.; Sohn, J.J.; Zanetti, K.A.; Bowman, E.D.; Yanaihara, N.; Yuen, S.T.; Chan, T.L.; Kwong, D.L.; Au, G.K.; et al. MicroRNA expression profiles associated with prognosis and therapeutic outcome in colon adenocarcinoma. JAMA 2008, 299, 425–436. [Google Scholar] [CrossRef]
- Volinia, S.; Calin, G.A.; Liu, C.G.; Ambs, S.; Cimmino, A.; Petrocca, F.; Visone, R.; Iorio, M.; Roldo, C.; Ferracin, M.; et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci. USA 2006, 103, 2257–2261. [Google Scholar] [CrossRef]
- Landgraf, P.; Rusu, M.; Sheridan, R.; Sewer, A.; Iovino, N.; Aravin, A.; Pfeffer, S.; Rice, A.; Kamphorst, A.O.; Landthaler, M.; et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 2007, 129, 1401–1414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciafrè, S.A.; Galardi, S.; Mangiola, A.; Ferracin, M.; Liu, C.G.; Sabatino, G.; Negrini, M.; Maira, G.; Croce, C.M.; Farace, M.G. Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem. Biophys. Res. Commun. 2005, 334, 1351–1358. [Google Scholar] [CrossRef] [PubMed]
- Li, R.; Qu, H.; Wang, S. CancerMIRNome: An interactive analysis and visualization database for miRNome profiles of human cancer. Nucleic Acids Res. 2022, 50, D1139–D1146. [Google Scholar] [CrossRef]
- Ren, Y.; Zhou, X.; Mei, M.; Yuan, X.B.; Han, L.; Wang, G.X.; Jia, Z.F.; Xu, P.; Pu, P.Y.; Kang, C.S. MicroRNA-21 inhibitor sensitizes human glioblastoma cells U251 (PTEN-mutant) and LN229 (PTEN-wild type) to taxol. BMC Cancer 2010, 10, 27. [Google Scholar] [CrossRef] [PubMed]
- Han, L.; Yue, X.; Zhou, X.; Lan, F.M.; You, G.; Zhang, W.; Zhang, K.L.; Zhang, C.Z.; Cheng, J.Q.; Yu, S.Z.; et al. MicroRNA-21 expression is regulated by β-catenin/STAT3 pathway and promotes glioma cell invasion by direct targeting RECK. CNS Neurosci. Ther. 2012, 18, 573–583. [Google Scholar] [CrossRef] [PubMed]
- Zhang, K.L.; Han, L.; Chen, L.Y.; Shi, Z.D.; Yang, M.; Ren, Y.; Chen, L.C.; Zhang, J.X.; Pu, P.Y.; Kang, C.S. Blockage of a miR-21/EGFR regulatory feedback loop augments anti-EGFR therapy in glioblastomas. Cancer Lett. 2014, 342, 139–149. [Google Scholar] [CrossRef] [PubMed]
- Kwak, H.J.; Kim, Y.J.; Chun, K.R.; Woo, Y.M.; Park, S.J.; Jeong, J.A.; Jo, S.H.; Kim, T.H.; Min, H.S.; Chae, J.S.; et al. Downregulation of Spry2 by miR-21 triggers malignancy in human gliomas. Oncogene 2011, 30, 2433–2442. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Q.; Chen, S.; Zhu, Z.; Yu, L.; Ren, Y.; Jiang, M.; Weng, J. miR-21 promotes EGF-induced pancreatic cancer cell proliferation by targeting Spry2. Cell Death Dis. 2018, 9, 1157. [Google Scholar] [CrossRef]
- Zhang, J.G.; Wang, J.J.; Zhao, F.; Liu, Q.; Jiang, K.; Yang, G.H. MicroRNA-21 (miR-21) represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer (NSCLC). Clin. Chim. Acta 2010, 411, 846–852. [Google Scholar] [CrossRef] [PubMed]
- Frankel, L.B.; Christoffersen, N.R.; Jacobsen, A.; Lindow, M.; Krogh, A.; Lund, A.H. Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J. Biol. Chem. 2008, 283, 1026–1033. [Google Scholar] [CrossRef]
- Zhu, S.; Wu, H.; Wu, F.; Nie, D.; Sheng, S.; Mo, Y.Y. MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res. 2008, 18, 350–359. [Google Scholar] [CrossRef] [Green Version]
- Medina, P.P.; Nolde, M.; Slack, F.J. OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature 2010, 467, 86–90. [Google Scholar] [CrossRef] [PubMed]
- Thomson, J.M.; Newman, M.; Parker, J.S.; Morin-Kensicki, E.M.; Wright, T.; Hammond, S.M. Extensive post-transcriptional regulation of microRNAs and its implications for cancer. Genes Dev. 2006, 20, 2202–2207. [Google Scholar] [CrossRef] [PubMed]
- Guil, S.; Cáceres, J.F. The multifunctional RNA-binding protein hnRNP A1 is required for processing of miR-18a. Nat. Struct. Mol. Biol. 2007, 14, 591–596. [Google Scholar] [CrossRef]
- Trabucchi, M.; Briata, P.; Garcia-Mayoral, M.; Haase, A.D.; Filipowicz, W.; Ramos, A.; Gherzi, R.; Rosenfeld, M.G. The RNA-binding protein KSRP promotes the biogenesis of a subset of microRNAs. Nature 2009, 459, 1010–1014. [Google Scholar] [CrossRef]
- Kim, Y.J.; Park, S.J.; Choi, E.Y.; Kim, S.; Kwak, H.J.; Yoo, B.C.; Yoo, H.; Lee, S.H.; Kim, D.; Park, J.B.; et al. PTEN modulates miR-21 processing via RNA-regulatory protein RNH1. PLoS ONE 2011, 6, e28308. [Google Scholar] [CrossRef]
- Wang, D.; Huang, J.; Hu, Z. RNA helicase DDX5 regulates microRNA expression and contributes to cytoskeletal reorganization in basal breast cancer cells. Mol. Cell Proteomics 2012, 11, M111.011932. [Google Scholar] [CrossRef]
- Yin, J.; Park, G.; Lee, J.E.; Choi, E.Y.; Park, J.Y.; Kim, T.H.; Park, N.; Jin, X.; Jung, J.E.; Shin, D.; et al. DEAD-box RNA helicase DDX23 modulates glioma malignancy via elevating miR-21 biogenesis. Brain 2015, 138, 2553–2570. [Google Scholar] [CrossRef]
- Zhang, Y.; Yu, F.; Ni, B.; Li, Q.; Bae, S.W.; Choi, J.H.; Yang, H.K.; Kong, S.H.; Zhu, C. The RNA-Binding Protein DDX18 Promotes Gastric Cancer by Affecting the Maturation of MicroRNA-21. Front. Oncol. 2020, 10, 598238. [Google Scholar] [CrossRef]
- Davis, B.N.; Hilyard, A.C.; Lagna, G.; Hata, A. SMAD proteins control DROSHA-mediated microRNA maturation. Nature 2008, 454, 56–61. [Google Scholar] [CrossRef]
- Telonis, A.G.; Loher, P.; Jing, Y.; Londin, E.; Rigoutsos, I. Beyond the one-locus-one-miRNA paradigm: MicroRNA isoforms enable deeper insights into breast cancer heterogeneity. Nucleic Acids Res. 2015, 43, 9158–9175. [Google Scholar] [CrossRef] [Green Version]
- Koi, Y.; Tsutani, Y. Predicting the presence of breast cancer using circulating small RNAs, including those in the extracellular vesicles. Cancer Sci. 2020, 111, 2104–2115. [Google Scholar] [CrossRef]
- Park, S.; Yang, H.D.; Seo, J.W.; Nam, J.W. hnRNPC induces isoform shifts in miR-21-5p leading to cancer development. Exp. Mol. Med. 2022, 54, 812–824. [Google Scholar] [CrossRef]
- Jiao, W.; Leng, X.; Zhou, Q.; Wu, Y.; Sun, L.; Tan, Y.; Ni, H.; Dong, X.; Shen, T.; Liu, Y. Different miR-21-3p isoforms and their different features in colorectal cancer. Int. J. Cancer 2017, 141, 2103–2111. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.C.; Skiados, N. MicroRNA-21 guide and passenger strand regulation of adenylosuccinate lyase-mediated purine metabolism promotes transition to an EGFR-TKI-tolerant persister state. Cancer Gene Ther. 2022. [Google Scholar] [CrossRef]
- Mortoglou, M.; Miralles, F. microRNA-21 Regulates Stemness in Pancreatic Ductal Adenocarcinoma Cells. Int. J. Mol. Sci. 2022, 23, 1275. [Google Scholar] [CrossRef] [PubMed]
- Yu, Y.; Kanwar, S.S.; Patel, B.B.; Oh, P.S.; Nautiyal, J.; Sarkar, F.H.; Majumdar, A.P. MicroRNA-21 induces stemness by downregulating transforming growth factor beta receptor 2 (TGFβR2) in colon cancer cells. Carcinogenesis 2012, 33, 68–76. [Google Scholar] [CrossRef]
- Caruso, S.; Bazan, V.; Rolfo, C.; Insalaco, L.; Fanale, D.; Bronte, G.; Corsini, L.R.; Rizzo, S.; Cicero, G.; Russo, A. MicroRNAs in colorectal cancer stem cells: New regulators of cancer stemness? Oncogenesis 2012, 1, e32. [Google Scholar] [CrossRef] [PubMed]
- Sathyan, P.; Zinn, P.O.; Marisetty, A.L.; Liu, B.; Kamal, M.M.; Singh, S.K.; Bady, P.; Lu, L.; Wani, K.M.; Veo, B.L.; et al. Mir-21-Sox2 Axis Delineates Glioblastoma Subtypes with Prognostic Impact. J. Neurosci. 2015, 35, 15097–15112. [Google Scholar] [CrossRef] [PubMed]
- Trohatou, O.; Zagoura, D.; Bitsika, V.; Pappa, K.I.; Antsaklis, A.; Anagnou, N.P.; Roubelakis, M.G. Sox2 suppression by miR-21 governs human mesenchymal stem cell properties. Stem Cells Transl. Med. 2014, 3, 54–68. [Google Scholar] [CrossRef]
- Hashimi, S.T.; Fulcher, J.A.; Chang, M.H.; Gov, L.; Wang, S.; Lee, B. MicroRNA profiling identifies miR-34a and miR-21 and their target genes JAG1 and WNT1 in the coordinate regulation of dendritic cell differentiation. Blood 2009, 114, 404–414. [Google Scholar] [CrossRef] [Green Version]
- Meng, F.; Henson, R.; Wehbe-Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef]
- Thum, T.; Gross, C.; Fiedler, J.; Fischer, T.; Kissler, S.; Bussen, M.; Galuppo, P.; Just, S.; Rottbauer, W.; Frantz, S.; et al. MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature 2008, 456, 980–984. [Google Scholar] [CrossRef] [PubMed]
- Gabriely, G.; Wurdinger, T.; Kesari, S.; Esau, C.C.; Burchard, J.; Linsley, P.S.; Krichevsky, A.M. MicroRNA 21 promotes glioma invasion by targeting matrix metalloproteinase regulators. Mol. Cell Biol. 2008, 28, 5369–5380. [Google Scholar] [CrossRef]
- Zhou, B.; Wang, D.; Sun, G.; Mei, F.; Cui, Y.; Xu, H. Effect of miR-21 on Apoptosis in Lung Cancer Cell Through Inhibiting the PI3K/ Akt/NF-κB Signaling Pathway in Vitro and in Vivo. Cell Physiol. Biochem. 2018, 46, 999–1008. [Google Scholar] [CrossRef] [PubMed]
- Afonso, M.B.; Rodrigues, P.M. miRNA-21 ablation protects against liver injury and necroptosis in cholestasis. Cell Death Differ. 2018, 25, 857–872. [Google Scholar] [CrossRef] [PubMed]
- Gong, Y.; Fan, Z.; Luo, G.; Yang, C.; Huang, Q.; Fan, K.; Cheng, H.; Jin, K.; Ni, Q.; Yu, X.; et al. The role of necroptosis in cancer biology and therapy. Mol. Cancer 2019, 18, 100. [Google Scholar] [CrossRef]
- Ma, X.; Conklin, D.J.; Li, F.; Dai, Z.; Hua, X.; Li, Y.; Xu-Monette, Z.Y.; Young, K.H.; Xiong, W.; Wysoczynski, M.; et al. The oncogenic microRNA miR-21 promotes regulated necrosis in mice. Nat. Commun. 2015, 6, 7151. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Peng, R.; Zeng, M.; Zhang, Z.; Liu, S.; Jiang, D.; Lu, Y. An autoregulatory feedback loop of miR-21/VMP1 is responsible for the abnormal expression of miR-21 in colorectal cancer cells. Cell Death Dis. 2020, 11, 1067. [Google Scholar] [CrossRef]
- He, C.; Dong, X.; Zhai, B.; Jiang, X.; Dong, D.; Li, B.; Jiang, H.; Xu, S.; Sun, X. MiR-21 mediates sorafenib resistance of hepatocellular carcinoma cells by inhibiting autophagy via the PTEN/Akt pathway. Oncotarget 2015, 6, 28867–28881. [Google Scholar] [CrossRef]
- Gwak, H.S.; Kim, T.H.; Jo, G.H.; Kim, Y.J.; Kwak, H.J.; Kim, J.H.; Yin, J.; Yoo, H.; Lee, S.H.; Park, J.B. Silencing of microRNA-21 confers radio-sensitivity through inhibition of the PI3K/AKT pathway and enhancing autophagy in malignant glioma cell lines. PLoS ONE 2012, 7, e47449. [Google Scholar] [CrossRef]
- Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
- Li, J.; Cao, F.; Yin, H.L.; Huang, Z.J.; Lin, Z.T.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, present and future. Cell Death Differ. 2020, 11, 88. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Wang, L.; Li, H.; Zhang, L.; Zheng, X.; Cheng, W. Crosstalk between noncoding RNAs and ferroptosis: New dawn for overcoming cancer progression. Cell Death Dis. 2020, 11, 580. [Google Scholar] [CrossRef] [PubMed]
- Guo, W.; Wu, Z.; Chen, J.; Guo, S.; You, W.; Wang, S.; Ma, J.; Wang, H.; Wang, X.; Wang, H.; et al. Nanoparticle delivery of miR-21-3p sensitizes melanoma to anti-PD-1 immunotherapy by promoting ferroptosis. J. Immunother. Cancer 2022, 10, e004381. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.X.; Lu, B.B.; Wang, H.; Cheng, Z.X.; Yin, Y.M. MicroRNA-21 modulates chemosensitivity of breast cancer cells to doxorubicin by targeting PTEN. Arch. Med. Res. 2011, 42, 281–290. [Google Scholar] [CrossRef]
- Yang, S.M.; Huang, C.; Li, X.F.; Yu, M.Z.; He, Y.; Li, J. miR-21 confers cisplatin resistance in gastric cancer cells by regulating PTEN. Toxicology 2013, 306, 162–168. [Google Scholar] [CrossRef] [PubMed]
- Dong, Z.; Ren, L.; Lin, L.; Li, J.; Huang, Y.; Li, J. Effect of microRNA-21 on multidrug resistance reversal in A549/DDP human lung cancer cells. Mol. Med. Rep. 2015, 11, 682–690. [Google Scholar] [CrossRef] [PubMed]
- Shi, L.; Chen, J.; Yang, J.; Pan, T.; Zhang, S.; Wang, Z. MiR-21 protected human glioblastoma U87MG cells from chemotherapeutic drug temozolomide induced apoptosis by decreasing Bax/Bcl-2 ratio and caspase-3 activity. Brain Res. 2010, 1352, 255–264. [Google Scholar] [CrossRef]
- Wang, P.; Zhuang, L.; Zhang, J.; Fan, J.; Luo, J.; Chen, H.; Wang, K.; Liu, L.; Chen, Z.; Meng, Z. The serum miR-21 level serves as a predictor for the chemosensitivity of advanced pancreatic cancer, and miR-21 expression confers chemoresistance by targeting FasL. Mol. Oncol. 2013, 7, 334–345. [Google Scholar] [CrossRef] [PubMed]
- Guraya, S. Prognostic significance of circulating microRNA-21 expression in esophageal, pancreatic and colorectal cancers; a systematic review and meta-analysis. Int. J. Surg. 2018, 60, 41–47. [Google Scholar] [CrossRef]
- Mace, T.A.; Collins, A.L.; Wojcik, S.E.; Croce, C.M.; Lesinski, G.B.; Bloomston, M. Hypoxia induces the overexpression of microRNA-21 in pancreatic cancer cells. J. Surg. Res. 2013, 184, 855–860. [Google Scholar] [CrossRef]
- Cortez, M.A.; Bueso-Ramos, C.; Ferdin, J.; Lopez-Berestein, G.; Sood, A.K.; Calin, G.A. MicroRNAs in body fluids--the mix of hormones and biomarkers. Nat. Rev. Clin. Oncol. 2011, 8, 467–477. [Google Scholar] [CrossRef]
- Creemers, E.E.; Tijsen, A.J.; Pinto, Y.M. Circulating microRNAs: Novel biomarkers and extracellular communicators in cardiovascular disease? Circ. Res. 2012, 110, 483–495. [Google Scholar] [CrossRef]
- Tomimaru, Y.; Eguchi, H.; Nagano, H.; Wada, H.; Kobayashi, S.; Marubashi, S.; Tanemura, M.; Tomokuni, A.; Takemasa, I.; Umeshita, K.; et al. Circulating microRNA-21 as a novel biomarker for hepatocellular carcinoma. J. Hepatol. 2012, 56, 167–175. [Google Scholar] [CrossRef]
- Sierzega, M.; Kaczor, M.; Kolodziejczyk, P.; Kulig, J.; Sanak, M.; Richter, P. Evaluation of serum microRNA biomarkers for gastric cancer based on blood and tissue pools profiling: The importance of miR-21 and miR-331. Br. J. Cancer 2017, 117, 266–273. [Google Scholar] [CrossRef]
- Guo, X.; Lv, X.; Lv, X.; Ma, Y.; Chen, L.; Chen, Y. Circulating miR-21 serves as a serum biomarker for hepatocellular carcinoma and correlated with distant metastasis. Oncotarget 2017, 8, 44050–44058. [Google Scholar] [CrossRef] [PubMed]
- Qu, K.Z.; Zhang, K.; Li, H.; Afdhal, N.H.; Albitar, M. Circulating microRNAs as biomarkers for hepatocellular carcinoma. J. Clin. Gastroenterol. 2011, 45, 355–360. [Google Scholar] [CrossRef]
- Lee, K.-Y.; Seo, Y.; Im, J.H.; Rhim, J.; Baek, W.; Kim, S.; Kwon, J.-W.; Yoo, B.C.; Shin, S.H.; Yoo, H.; et al. Molecular Signature of Extracellular Vesicular Small Non-Coding RNAs Derived from Cerebrospinal Fluid of Leptomeningeal Metastasis Patients: Functional Implication of miR-21 and Other Small RNAs in Cancer Malignancy. Cancers 2021, 13, 209. [Google Scholar] [CrossRef] [PubMed]
- Im, J.H.; Yoo, B.C.; Lee, J.H.; Lee, K.-Y.; Kim, K.-H.; Kim, J.H.; Park, H.J.; Park, M.; Lee, S.H.; Kwon, J.-W.; et al. Experimental Assessment of Leptomeningeal Metastasis Diagnosis in Medulloblastoma Using Cerebrospinal Fluid Metabolomic Profiles. Metabolites 2021, 11, 851. [Google Scholar] [CrossRef]
- Toiyama, Y.; Takahashi, M.; Hur, K.; Nagasaka, T.; Tanaka, K.; Inoue, Y.; Kusunoki, M.; Boland, C.R.; Goel, A. Serum miR-21 as a diagnostic and prognostic biomarker in colorectal cancer. J. Natl. Cancer Inst. 2013, 105, 849–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taylor, D.D.; Gercel-Taylor, C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2008, 110, 13–21. [Google Scholar] [CrossRef] [PubMed]
- Lee, K.Y.; Im, J.H.; Lin, W.; Gwak, H.S. Nanoparticles in 472 Human Cerebrospinal Fluid: Changes in Extracellular Vesicle Concentration and miR-21 Expression as a Biomarker for Leptomeningeal Metastasis. Cancers 2020, 12, 2745. [Google Scholar] [CrossRef]
- Im, J.H.; Kim, T.H.; Lee, K.-Y.; Gwak, H.-S.; Lin, W.; Park, J.B.; Kim, J.H.; Yoo, B.C.; Park, S.-M.; Kwon, J.-W.; et al. Exploratory Profiling of Extracellular MicroRNAs in Cerebrospinal Fluid Comparing Leptomeningeal Metastasis with Other Central Nervous System Tumor Statuses. J. Clin. Med. 2021, 10, 4860. [Google Scholar] [CrossRef] [PubMed]
- Im, J.H.; Yoo, B.C.; Lee, J.H.; Kim, K.H.; Kim, T.H.; Lee, K.Y.; Kim, J.H.; Park, J.B.; Kwon, J.W.; Shin, S.H.; et al. Comparative cerebrospinal fluid metabolites profiling in glioma patients to predict malignant transformation and leptomeningeal metastasis with a potential for preventive personalized medicine. EPMA J. 2020, 11, 469–484. [Google Scholar] [CrossRef] [PubMed]
- Li, J.R.; Tong, C.Y.; Sung, T.J.; Kang, T.Y.; Zhou, X.J.; Liu, C.C. CMEP: A database for circulating microRNA expression profiling. Bioinformatics 2019, 35, 3127–3132. [Google Scholar] [CrossRef] [PubMed]
- Oughtred, R.; Rust, J.; Chang, C.; Breitkreutz, B.J.; Stark, C.; Willems, A.; Boucher, L.; Leung, G.; Kolas, N.; Zhang, F.; et al. The BioGRID database: A comprehensive biomedical resource of curated protein, genetic, and chemical interactions. Protein Sci. 2021, 30, 187–200. [Google Scholar] [CrossRef] [PubMed]
- Stark, C.; Breitkreutz, B.J.; Reguly, T.; Boucher, L.; Breitkreutz, A.; Tyers, M. BioGRID: A general repository for interaction datasets. Nucleic Acids Res. 2006, 34, D535–D539. [Google Scholar] [CrossRef] [PubMed]
- Jeggari, A.; Marks, D.S.; Larsson, E. miRcode: A map of putative microRNA target sites in the long non-coding transcriptome. Bioinformatics 2012, 28, 2062–2063. [Google Scholar] [CrossRef] [PubMed]
- Seo, Y.; Kim, S.S.; Kim, N.; Cho, S. Development of a miRNA-controlled dual-sensing system and its application for targeting miR-21 signaling in tumorigenesis. Exp. Mol. Med. 2020, 52, 1989–2004. [Google Scholar] [CrossRef]
- Nedaeinia, R.; Sharifi, M. Locked nucleic acid anti-miR-21 inhibits cell growth and invasive behaviors of a colorectal adenocarcinoma cell line: LNA-anti-miR as a novel approach. Cancer Gene Ther. 2016, 23, 246–253. [Google Scholar] [CrossRef] [PubMed]
- Javanmard, S.H.; Vaseghi, G.; Ghasemi, A.; Rafiee, L.; Ferns, G.A.; Esfahani, H.N.; Nedaeinia, R. Therapeutic inhibition of microRNA-21 (miR-21) using locked-nucleic acid (LNA)-anti-miR and its effects on the biological behaviors of melanoma cancer cells in preclinical studies. Cancer Cell Int. 2020, 20, 384. [Google Scholar] [CrossRef] [PubMed]
- Lee, T.J.; Yoo, J.Y.; Shu, D.; Li, H.; Zhang, J.; Yu, J.G.; Jaime-Ramirez, A.C.; Acunzo, M.; Romano, G.; Cui, R.; et al. RNA Nanoparticle-Based Targeted Therapy for Glioblastoma through Inhibition of Oncogenic miR-21. Mol. Ther. 2017, 25, 1544–1555. [Google Scholar] [CrossRef] [PubMed]
- Griveau, A.; Bejaud, J.; Anthiya, S.; Avril, S.; Autret, D.; Garcion, E. Silencing of miR-21 by locked nucleic acid-lipid nanocapsule complexes sensitize human glioblastoma cells to radiation-induced cell death. Int. J. Pharm. 2013, 454, 765–774. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; Huang, Y.; Chen, D.; He, J.; Zhu, W.; Zhang, Y.; Liu, X. Downregulation of miR-21 increases cisplatin sensitivity of non-small-cell lung cancer. Cancer Genet. 2014, 207, 214–220. [Google Scholar] [CrossRef]
- Ren, J.; Zhu, D.; Liu, M.; Sun, Y.; Tian, L. Downregulation of miR-21 modulates Ras expression to promote apoptosis and suppress invasion of Laryngeal squamous cell carcinoma. Eur. J. Cancer 2010, 46, 3409–3416. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Ren, S.; Li, X.; Wang, Y.; Garfield, D.; Zhou, S.; Chen, X.; Su, C.; Chen, M.; Kuang, P.; et al. MiR-21 overexpression is associated with acquired resistance of EGFR-TKI in non-small cell lung cancer. Lung Cancer 2014, 83, 146–153. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Li, W.; Yang, Y.; Lu, Y.; He, C.; Hu, G.; Liu, H.; Chen, J.; He, J.; Yu, H. MicroRNA-21 targets LRRFIP1 and contributes to VM-26 resistance in glioblastoma multiforme. Brain Res. 2009, 1286, 13–18. [Google Scholar] [CrossRef] [PubMed]
- Giunti, L.; da Ros, M.; Vinci, S.; Gelmini, S.; Iorio, A.L.; Buccoliero, A.M.; Cardellicchio, S.; Castiglione, F.; Genitori, L.; de Martino, M.; et al. Anti-miR21 oligonucleotide enhances chemosensitivity of T98G cell line to doxorubicin by inducing apoptosis. Am. J. Cancer Res. 2015, 5, 231–242. [Google Scholar] [PubMed]
- Connolly, E.C.; Van Doorslaer, K.; Rogler, L.E.; Rogler, C.E. Overexpression of miR-21 promotes an in vitro metastatic phenotype by targeting the tumor suppressor RHOB. Mol. Cancer Res. 2010, 8, 691–700. [Google Scholar] [CrossRef]
- Gong, C.; Nie, Y.; Qu, S.; Liao, J.Y.; Cui, X.; Yao, H.; Zeng, Y.; Su, F.; Song, E.; Liu, Q. miR-21 induces myofibroblast differentiation and promotes the malignant progression of breast phyllodes tumors. Cancer Res. 2014, 74, 4341–4352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, X.; Abraham, J.M.; Cheng, Y.; Wang, Z.; Wang, Z.; Zhang, G.; Ashktorab, H.; Smoot, D.T.; Cole, R.N.; Boronina, T.N.; et al. Synthetic Circular RNA Functions as a miR-21 Sponge to Suppress Gastric Carcinoma Cell Proliferation. Mol. Ther. Nucleic Acids 2018, 13, 312–321. [Google Scholar] [CrossRef]
- Rama, A.R.; Quiñonero, F. Synthetic Circular miR-21 Sponge as Tool for Lung Cancer Treatment. Int. J. Mol. Sci. 2022, 23, 2963. [Google Scholar] [CrossRef] [PubMed]
- Davis, S.; Lollo, B.; Freier, S.; Esau, C. Improved targeting of miRNA with antisense oligonucleotides. Nucleic Acids Res. 2006, 34, 2294–2304. [Google Scholar] [CrossRef]
- Burris, H.A., 3rd; Moore, M.J.; Andersen, J.; Green, M.R.; Rothenberg, M.L.; Modiano, M.R.; Cripps, M.C.; Portenoy, R.K.; Storniolo, A.M.; Tarassoff, P.; et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J. Clin. Oncol. 1997, 15, 2403–2413. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Chen, Y.; Li, J.; Zhang, Z.; Huang, C.; Lian, G.; Yang, K.; Chen, S.; Lin, Y.; Wang, L.; et al. Co-delivery of microRNA-21 antisense oligonucleotides and gemcitabine using nanomedicine for pancreatic cancer therapy. Cancer Sci. 2017, 108, 1493–1503. [Google Scholar] [CrossRef] [PubMed]
- Si, M.L.; Zhu, S.; Wu, H.; Lu, Z.; Wu, F.; Mo, Y.Y. miR-21-mediated tumor growth. Oncogene 2007, 26, 2799–2803. [Google Scholar] [CrossRef]
- Leone, E.; Morelli, E.; Di Martino, M.T.; Amodio, N.; Foresta, U.; Gullà, A.; Rossi, M.; Neri, A.; Giordano, A.; Munshi, N.C.; et al. Targeting miR-21 inhibits in vitro and in vivo multiple myeloma cell growth. Clin. Cancer Res. 2013, 19, 2096–2106. [Google Scholar] [CrossRef] [PubMed]
- Wagenaar, T.R.; Tolstykh, T.; Shi, C.; Jiang, L.; Zhang, J.; Li, Z.; Yu, Q.; Qu, H.; Sun, F.; Cao, H.; et al. Identification of the endosomal sorting complex required for transport-I (ESCRT-I) as an important modulator of anti-miR uptake by cancer cells. Nucleic Acids Res. 2015, 43, 1204–1215. [Google Scholar] [CrossRef]
- Zhou, X.; Zhang, J.; Jia, Q.; Ren, Y.; Wang, Y.; Shi, L.; Liu, N.; Wang, G.; Pu, P.; You, Y.; et al. Reduction of miR-21 induces glioma cell apoptosis via activating caspase 9 and 3. Oncol. Rep. 2010, 24, 195–201. [Google Scholar] [CrossRef] [PubMed]
- Koshkin, A.A.; Singh, S.K.; Nielsen, P.; Rajwanshi, V.K.; Kumar, R.; Meldgaard, M.; Olsen, C.E.; Wengel, J. LNA (Locked Nucleic Acids): Synthesis of the adenine, cytosine, guanine, 5-methylcytosine, thymine and uracil bicyclonucleoside monomers, oligomerisation, and unprecedented nucleic acid recognition. Tetrahedron 1998, 54, 3607–3630. [Google Scholar] [CrossRef]
- Lima, J.F.; Cerqueira, L. Anti-miRNA oligonucleotides: A comprehensive guide for design. RNA Biol. 2018, 15, 338–352. [Google Scholar] [CrossRef] [PubMed]
- Obad, S.; dos Santos, C.O.; Petri, A.; Heidenblad, M.; Broom, O.; Ruse, C.; Fu, C.; Lindow, M.; Stenvang, J.; Straarup, E.M.; et al. Silencing of microRNA families by seed-targeting tiny LNAs. Nat. Genet. 2011, 43, 371–378. [Google Scholar] [CrossRef] [PubMed]
- Du, W.W.; Zhang, C.; Yang, W.; Yong, T.; Awan, F.M.; Yang, B.B. Identifying and Characterizing circRNA-Protein Interaction. Theranostics 2017, 7, 4183–4191. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Huang, C.; Bao, C.; Chen, L.; Lin, M.; Wang, X.; Zhong, G.; Yu, B.; Hu, W.; Dai, L.; et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat. Struct. Mol. Biol. 2015, 22, 256–264. [Google Scholar] [CrossRef]
- Zhong, Y.; Du, Y.; Yang, X.; Mo, Y.; Fan, C.; Xiong, F.; Ren, D.; Ye, X.; Li, C.; Wang, Y.; et al. Circular RNAs function as ceRNAs to regulate and control human cancer progression. Mol. Cancer 2018, 17, 79. [Google Scholar] [CrossRef]
- Anastasiadou, E.; Jacob, L.S.; Slack, F.J. Non-coding RNA networks in cancer. Nat. Rev. Cancer 2018, 18, 5–18. [Google Scholar] [CrossRef] [PubMed]
- Ebert, M.S.; Neilson, J.R.; Sharp, P.A. MicroRNA sponges: Competitive inhibitors of small RNAs in mammalian cells. Nat. Methods 2007, 4, 721–726. [Google Scholar] [CrossRef] [PubMed]
- Jung, J.; Yeom, C.; Choi, Y.S.; Kim, S.; Lee, E.; Park, M.J.; Kang, S.W.; Kim, S.B.; Chang, S. Simultaneous inhibition of multiple oncogenic miRNAs by a multi-potent microRNA sponge. Oncotarget 2015, 6, 20370–20387. [Google Scholar] [CrossRef]
- Hansen, T.B.; Jensen, T.I.; Clausen, B.H.; Bramsen, J.B.; Finsen, B.; Damgaard, C.K.; Kjems, J. Natural RNA circles function as efficient microRNA sponges. Nature 2013, 495, 384–388. [Google Scholar] [CrossRef]
- Zhang, C.C.; Li, Y.; Feng, X.Z.; Li, D.B. Circular RNA circ_0001287 inhibits the proliferation, metastasis, and radiosensitivity of non-small cell lung cancer cells by sponging microRNA miR-21 and up-regulating phosphatase and tensin homolog expression. Bioengineered 2021, 12, 414–425. [Google Scholar] [CrossRef]
- Müller, S.; Wedler, A.; Breuer, J.; Glaß, M.; Bley, N.; Lederer, M.; Haase, J.; Misiak, C.; Fuchs, T.; Ottmann, A.; et al. Synthetic circular miR-21 RNA decoys enhance tumor suppressor expression and impair tumor growth in mice. NAR Cancer 2020, 2, zcaa014. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Yang, T.; Yu, Z.; Liu, T.; Jin, R.; Weng, L.; Bai, Y.; Gooding, J.J.; Zhang, Y.; Chen, X. Intelligent Gold Nanoparticles with Oncogenic MicroRNA-Dependent Activities to Manipulate Tumorigenic Environments for Synergistic Tumor Therapy. Adv. Mater. 2022, 34, e2110219. [Google Scholar] [CrossRef] [PubMed]
- Winkle, M.; El-Daly, S.M.; Fabbri, M. Noncoding RNA therapeutics—Challenges and potential solutions. Nat. Rev. Drug Discov. 2021, 20, 629–651. [Google Scholar] [CrossRef]
- Gumireddy, K.; Young, D.D.; Xiong, X.; Hogenesch, J.B.; Huang, Q.; Deiters, A. Small-molecule inhibitors of microrna miR-21 function. Angew. Chem. Int. Ed. Engl. 2008, 47, 7482–7484. [Google Scholar] [CrossRef]
- Wickramasinghe, N.S.; Manavalan, T.T.; Dougherty, S.M.; Riggs, K.A.; Li, Y.; Klinge, C.M. Estradiol downregulates miR-21 expression and increases miR-21 target gene expression in MCF-7 breast cancer cells. Nucleic Acids Res. 2009, 37, 2584–2595. [Google Scholar] [CrossRef] [PubMed]
- Fu, Z.; Wang, L.; Li, S.; Chen, F.; Au-Yeung, K.K.; Shi, C. MicroRNA as an Important Target for Anticancer Drug Development. Front. Pharmacol. 2021, 12, 736323. [Google Scholar] [CrossRef]
- Song, J.H.; Kang, H.J.; Luevano, L.A.; Gokhale, V.; Wu, K.; Pandey, R.; Sherry Chow, H.H.; Hurley, L.H.; Kraft, A.S. Small-Molecule-Targeting Hairpin Loop of hTERT Promoter G-Quadruplex Induces Cancer Cell Death. Cell Chem Biol. 2019, 26, 1110–1121. [Google Scholar] [CrossRef] [PubMed]
Subtype | Abbreviation | Size (nt) | Description | References |
---|---|---|---|---|
Circular RNA | circRNA | 100 ~ ≥4000 | Functions through interaction with RNA and RNA binding proteins; modulating stability and regulating gene transcription | [23,24,25] |
Enhancer RNA | eRNA | ≤2000 | Transcribed upon activation of enhancers and affects efficiency of enhancer activation and gene transcription | [26,27] |
Piwi-interacting RNA | piRNA | 26–31 | Forms RNA-protein complexes with Piwi-subfamily proteins and involving in epigenetic and post- transcriptional silencing of transposable elements | [28] |
Small interfering RNA | siRNA | 21–25 | Plays a role in RNA interference through silencing of transposons and mediating transcriptional gene silencing | [29] |
Small nuclear RNA | snRNA | ~150 | Primarily functions in processing of pre-mRNA or heterogeneous nuclear RNA (hnRNA) in the nucleus | [30] |
Small nucleolar RNA | snoRNA | ~70–250 | Primarily guide chemical modification and processing of other RNAs and also function as miRNA | [31] |
Telomerase RNA component | TERC | 150–1300 | Helps constitute telomerase activity in tandem with TERT and acts as a template for telomere replication | [32] |
tRNA-derived small RNA | tsRNA | 18–40 | Regulates gene expression similarly to miRNA- mediated RNA silencing mechanism via interacting with Ago proteins | [33] |
Vault RNA | vtRNA | 86–141 | Associated with drug resistance and regulates expression through miRNA-like RNA silencing mechanism via interacting with Ago proteins | [34] |
Y RNA | Y RNA | 84–113 | Components of Ro60 ribonucleoprotein and involved in various cellular processes such as DNA replication and RNA quality control | [35,36] |
Type | Target Cancers | Cell Lines & Experimental Models | Phenotypes | References |
---|---|---|---|---|
LNA | Colorectal adenocarcinoma | LS174T | Proliferation inhibited; apoptosis enhanced | [120] |
Melanoma | B16F10; C57BL/6 mice | Tumor growth and volume inhibited; apoptosis enhanced | [121] | |
Glioblastoma | U87MG; Orthotopic xenograft in athymic nude mice | Tumor growth inhibited; apoptosis enhanced | [122,123] | |
Non-small cell lung cancer | A549; Female nude mice | Drug sensitization; tumor growth inhibited; apoptosis enhanced | [124] | |
Breast cancer | MCF-7 | Proliferation inhibited | [59] | |
ASO | Laryngeal squamous cell carcinoma | Hep-2; BALB/c nude mice | Tumor growth and proliferation inhibited; invasiveness decreased; cell cycle arrest; apoptosis enhanced | [125] |
Non-small cell lung cancer | PC9; Female BALB/c nude mice | Proliferation inhibited; apoptosis enhanced; tumor growth inhibited | [126] | |
Glioblastoma multiforme | LN229; U251MG; U373MG; T98G | Drug sensitization; cell viability decreased | [127,128] | |
Hepatocellular carcinoma | Huh7; HepG2 | Migration and invasiveness decreased | [129] | |
Breast phyllode tumor | Patient-derived breast stromal cells; Female nude mice | Proliferation inhibited and invasion decreased | [130] | |
CircRNA | Gastric carcinoma | NCI-N87; AGS; MKN28 | Proliferation inhibited | [131] |
Lung cancer | L132, A549; LL2; 3D multicellular spheroids | Proliferation inhibited; migration decreased; apoptosis enhanced | [132] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Rhim, J.; Baek, W.; Seo, Y.; Kim, J.H. From Molecular Mechanisms to Therapeutics: Understanding MicroRNA-21 in Cancer. Cells 2022, 11, 2791. https://doi.org/10.3390/cells11182791
Rhim J, Baek W, Seo Y, Kim JH. From Molecular Mechanisms to Therapeutics: Understanding MicroRNA-21 in Cancer. Cells. 2022; 11(18):2791. https://doi.org/10.3390/cells11182791
Chicago/Turabian StyleRhim, Jiho, Woosun Baek, Yoona Seo, and Jong Heon Kim. 2022. "From Molecular Mechanisms to Therapeutics: Understanding MicroRNA-21 in Cancer" Cells 11, no. 18: 2791. https://doi.org/10.3390/cells11182791
APA StyleRhim, J., Baek, W., Seo, Y., & Kim, J. H. (2022). From Molecular Mechanisms to Therapeutics: Understanding MicroRNA-21 in Cancer. Cells, 11(18), 2791. https://doi.org/10.3390/cells11182791