miRNA Dysregulation in Cancer: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor:

miRNAs are a subset of non-coding RNAs that regulate the expression of a multitude of genes post-transcriptionally and thus are potential diagnostic, prognostic, and predictive biomarkers and have also emerged as potential therapeutics. Because miRNAs are involved in the post-transcriptional regulation of their target mRNAs via repressing gene expression, defects in miRNA biogenesis pathway and miRNA expression perturb the expression of a multitude of oncogenic or tumor-suppressive genes that are involved in the pathogenesis of various cancers. As such, numerous miRNAs have been identified to be downregulated or upregulated in many cancers, functioning as either oncomes or oncosuppressor miRs. Moreover, dysregulation of miRNA biogenesis pathways can also change miRNA expression and function in cancer.

  • microRNAs
  • circulating miRNAs
  • post-transcriptional gene regulation

1. microRNAs

After the discovery of the first microRNA (miRNA), lin-4, in 1993 in Caenorhabditis elegans [1][2], it was quickly recognized that microRNAs have been detected throughout the animal and plant kingdoms and some were shown to be highly conserved across species [3][4][5], with a conserved mechanism and broad functional significance.
MiRNAs are small non-coding RNAs, with an average of 22 nucleotides in length which are highly conserved and are naturally encoded in the genomes of various species [3][4][5][6] and play key functional roles in the regulation of gene expression at the transcriptional [7][8][9] and post-transcriptional [10][11][12][13][14] levels of their target mRNAs [10][12] including the regulation of cell function by regulating gene expression via the modulation of the stability and translation of mRNA [15] in a broad range of biological processes within cells and organisms, consequently affecting cell differentiation, cell proliferation, angiogenesis, and apoptosis, etc. [16]. In addition, miRNAs exhibit tissue-specific [17][18] and developmental expression patterns [19][20][21].
Emerging data suggest that new miRNAs are still being discovered [22] and curreently, it is estimated that there are >2588 human mature miRNAs in human cells with time- and tissue-dependent expression patterns. Of the >2588 miRNAs, 1115 are currently annotated in miRBase V22 [23][24][25][26][27][28][29]. It is estimated that these >2588 miRNAs regulate over 60% of the expression of human genes, demonstrating that they have important regulatory roles in diverse physiological and developmental processes as well as in the pathogenesis of various human diseases and disorders [10][25][30][31][32][33][34][35][36][37][38][39][40][41][42][43][44][45][46][47][48][49][50][51], including many cancers [16][17][52][53][54][55][56][57][58][59][60][61][62][63][64][65][66][67][68][69][70][71][72][73][74][75][76][77][78][79][80].
As illustrated in Figure 1, most miRNAs are transcribed from DNA sequences into nascent primary miRNAs (pri-miRNAs) which are then initially processed by DROSHA in the nucleus to generate precursor miRNA (pre-miRNA) [10][81]. As many as 40% of miRNA genes may lie in the introns or even exons of other genes [82]. Pre-miRNAs are subsequently exported from the nucleus to the cytoplasm by exportin 5 (XPO5) where they are further processed by DICER, producing small RNA duplexes with 2 nt. 3′ overhangs. These small double-stranded RNA duplexes with distinct 2 nucleotides 3′ overhangs are then loaded onto the Argonaute (AGO) protein, which retains only one strand of mature miRNA by removing the other strand [12]. The complex formed by the AGO and miRNA associates with cofactors such as GW182 (i.e., TNRC6A), and forms the effector complex, the RNA-induced silencing complex (RISC) [14]. The miRNA-RISC complex is responsible for the degradation of mRNA transcripts and translational suppression through interaction with the complementary mRNA target sequences predominantly located within the 3′-untranslated region (3′-UTR) of mRNAs (Figure 1) [83][84][85][86].
Figure 1. Schematic of canonical and noncanonical miRNA biogenesis, processing, and target RNA translational suppression or degradation. The canonical miRNA pathway generates pri-miRNAs from transcripts by miRNA genes encoded in intronic, exonic, or intergenic regions. The pri-miRNA transcripts are then processed by Drosha/DGCR8 into pre-miRNAs. Intronic pre-miRNAs of the noncanonical mirtron pathway are formed by splicing, debranching, and trimming short introns without the involvement of Drosha processing. The pre-miRNA transcripts produced by both the canonical and noncanonical pathways are then exported from the nucleus to the cytoplasm by Exportin 5. This is followed by subsequent Dicer cleavage within the RISC loading complex (RLC) and subsequent unwinding of the miRNA/miRNA duplex via Argonaute, and TRBP-dependent loading into the RNA-induced silencing complex (RISC). The binding of target mRNAs to miRNAs in RISC is followed by suppression of translation and/or mRNA degradation within p-bodies in the cytosol. Created with BioRender.com (accessed on 6 July 2023). Abbreviations: AGO2: Argonaute RISC Catalytic Component 2; DICER1: Dicer 1, Ribonuclease III; EGFR: Epidermal growth factor receptor 2; DGCR8: DiGeorge Syndrome Critical Region 8; DROSHA: Drosha Ribonuclease III; mRNA: messenger RNA; miRNA: micro RNA; P-bodies: Processing bodies; pre-miRNA: precursor microRNA; pri-miRNA: primary microRNA; RNA Pol II: RNA polymerase II; TRBP: AR RNA-binding protein; XPO5: Exportin 5.
As discussed in the literature [67], a specific miRNA may target many different mRNAs [87]; while a particular messenger RNA may be targeted by several miRNAs, either simultaneously or in a context-dependent fashion [88], resulting in the cooperative repression effect [89][90].

2. miRNA Dysregulation in Cancer

Cancer is a complex and heterogeneous disease that evolves through successive genetic and genomic changes that support tumorigenesis [91]. Changes in the genome that affect gene function often result from genomic alterations such as chromosomal translocations, insertions or deletions, amplifications, and single-nucleotide mutations in the epigenome. These genetic and epigenetic alterations often trigger the activation of oncogenes and suppression of tumor suppressor genes [92]. To add to this picture, miRNAs also play an important role in organismal development, normal physiology, and disease state, including many hallmarks of various cancer types (Figure 2).
Figure 2. miRNAs play key roles at various stages of cancer development. Created with BioRender.com (accessed on 6 July 2023). Abbreviations: ca: cancer.
Although genetic, genomic, and epigenetic changes affecting genome function as well as the dysfunctions of various types of regulators are the primary molecular mechanisms responsible for the pathological processes of a multitude of human diseases, including many cancers, among which, miRNAs and their altered expression, and alterations and mutations in members of miRNA biogenesis pathways have also been recognized as additional molecular mechanisms implicated in the pathological processes of many cancers and have received great attention in recent years. Numerous studies have demonstrated the involvement of miRNAs in cancer pathogenesis by regulating the expression of their target mRNAs contributing to tumor growth, invasion, angiogenesis, and immune system evasion [93][94]. Furthermore, studies have shown that tumor-specific miRNA signatures can define cancer subtypes, patient survival, and treatment response [55][95][96], and notably, cancer-associated miRNAs can be detected in biological fluids, allowing less-invasive monitoring [97].
It is widely accepted that genetic deletion or amplification, epigenetic methylation of miRNA genomic loci, and alterations affecting the transcription factor-mediated regulation of primary miRNA (pri-miRNA) as well as factors involved in the miRNA biogenesis pathway frequently alter miRNA expression and function in many cancers.
Alterations throughout miRNA biogenesis can affect the availability of target mRNA, including many mRNAs associated with cancer development (Figure 3). As such, altered or dysregulated miRNAs or miRNA-processing machinery have been linked to many pathological processes, resulting in the failure to maintain the normal homeostatic state and eventually leading to malignant transformation, including many cancers [16][17][52][53][54][55][56][57][58][59][60][61][62][63][64][65][66][67][68][69][70][71][72][73][74][75][76][77][78][79][80].
Figure 3. Alterations throughout miRNA biogenesis can affect the availability of target mRNA, including many mRNAs associated with cancer development. The figure also depicts several modulators, proteins such as EGFR, and transcription factors such as ETS1/ELK1, MYC, epigenetic regulators such as demethylating protein KDM6A, a known tumor suppressor, the tumor suppressor gene TAP63, and miRNAs such as miR-103 that may interact with modulators of miRNA-processing machinery. Both KRAS and EGFR are essential mediators of pancreatic cancer development and interact with AGO2 to perturb its function. Created with BioRender.com (accessed on 6 July 2023). Abbreviations: AGO2: Argonaute RISC Catalytic Component 2; DICER1: Dicer 1, Ribonuclease III; DROSHA: Drosha Ribonuclease III; EGFR: Epidermal growth factor receptor 2; EGFR: Epidermal growth factor receptor, ELK1: ETS Like-1 protein Elk-1; ETS1: ETS Proto-Oncogene 1; KDM6A: Lysine (K)-specific demethylase 6A; miRNA: micro RNA; mRNA: messenger RNA; MYC: MYC Proto-Oncogene; pre-miRNA: precursor microRNA; pri-miRNA: primary microRNA; RNA Pol II: RNA polymerase II; TAP63: Tumor protein p63; XPO5: Exportin 5.
As illustrated in Figure 3, various genetic or epigenetic alterations in key factors involving the miRNA biogenesis process can affect the availability of miRNA targets, including many targets associated with cancer development, as well as several modulators and proteins (e.g., EGFR), transcription factors (e.g., ETS1/ELK1, MYC), epigenetic regulators (e.g., demethylating protein KDM6A, a known tumor suppressor), the tumor suppressor gene TAP63, and miRNAs (e.g., miR-103) [98] that may interact with factors involved in miRNA biogenesis machinery. It has been shown that both KRAS and EGFR are essential mediators of pancreatic cancer development and they were shown to interact with Argonaute 2 (AGO2) to perturb its function [99].
Recent findings indicate that different types of miRNAs have been shown to regulate many of the hallmarks of cancer Figure 2 [78], either as a tumor suppressor (i.e., oncosuppressor miRs) or as oncogenes (i.e., oncomirs) [100][101][102][103][104][105][106][107][108][109][110]. MiRNAs play a critical role in the regulation of the expression of a multitude of genes, such as the cellular responses to environmental stresses including DNA damage, hypoxia, oxidative stress, and starvation.
Supporting this, miRNAs with an oncogenic function as well as tumor-suppressing function have recently been identified in various neoplastic malignancies, and dysregulation of miRNA expression is closely associated with cancer initiation, progression, and metastasis and shown to be associated with the origin, progression, therapeutic response, and patient survival of the disease [17][56][59][63][70][79][80][111][112].
For example, the tissue specificity of miRNAs [17][18], which is required for maintaining normal cell and tissue homeostasis [55], allows them to be used as potential biomarkers in diagnosing cancer of unknown primary origin [28][113].
Supporting these findings, recurrent genetic and epigenetic alterations of individual miRNAs and components involved in the miRNA-processing machinery and biogenesis pathway have been identified in many cancer types, and emerging data from several functional studies provide evidence for the mechanism of action of many potential oncogenic and tumor-suppressor miRNAs [100][101][102][103][104][105][106][107][108][109][110] with potential clinical applications as potential diagnostics and therapeutics.

This entry is adapted from the peer-reviewed paper 10.3390/ijms241713340

References

  1. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854.
  2. Wightman, B.; Ha, I.; Ruvkun, G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993, 75, 855–862.
  3. Pasquinelli, A.E.; Reinhart, B.J.; Slack, F.; Martindale, M.Q.; Kuroda, M.I.; Maller, B.; Hayward, D.C.; Ball, E.E.; Degnan, B.; Müller, P.; et al. Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature 2000, 408, 86–89.
  4. Li, S.C.; Chan, W.C.; Hu, L.Y.; Lai, C.H.; Hsu, C.N.; Lin, W.C. Identification of homologous microRNAs in 56 animal genomes. Genomics 2010, 96, 1–9.
  5. Friedländer, M.R.; Lizano, E.; Houben, A.J.S.; Bezdan, D.; Báñez-Coronel, M.; Kudla, G.; Mateu-Huertas, E.; Kagerbauer, B.; González, J.; Chen, K.C.; et al. Evidence for the biogenesis of more than 1000 novel human microRNAs. Genome Biol. 2014, 15, R57.
  6. Griffiths-Jones, S.; Grocock, R.J.; van Dongen, S.; Bateman, A.; Enright, A.J. miRBase: MicroRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006, 34, D140–D144.
  7. Khraiwesh, B.; Arif, M.A.; Seumel, G.I.; Ossowski, S.; Weigel, D.; Reski, R.; Frank, W. Transcriptional control of gene expression by microRNAs. Cell 2010, 140, 111–122.
  8. Samad, A.F.A.; Sajad, M.; Nazaruddin, N.; Fauzi, I.A.; Murad, A.M.A.; Zainal, Z.; Ismail, I. MicroRNA and Transcription Factor: Key Players in Plant Regulatory Network. Front. Plant Sci. 2017, 8, 565.
  9. Tong, Z.; Cui, Q.; Wang, J.; Zhou, Y. TransmiR v2.0: An updated transcription factor-microRNA regulation database. Nucleic Acids Res. 2018, 47, D253–D258.
  10. Seyhan, A.A. microRNAs with different functions and roles in disease development and as potential biomarkers of diabetes: Progress and challenges. Mol. Biosyst. 2015, 11, 1217–1234.
  11. Chen, P.Y.; Meister, G. microRNA-guided posttranscriptional gene regulation. Biol. Chem. 2005, 386, 1205–1218.
  12. Catalanotto, C.; Cogoni, C.; Zardo, G. MicroRNA in Control of Gene Expression: An Overview of Nuclear Functions. Int. J. Mol. Sci. 2016, 17, 1712.
  13. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402.
  14. Pu, M.; Chen, J.; Tao, Z.; Miao, L.; Qi, X.; Wang, Y.; Ren, J. Regulatory network of miRNA on its target: Coordination between transcriptional and post-transcriptional regulation of gene expression. Cell. Mol. Life Sci. 2019, 76, 441–451.
  15. van Rooij, E. The art of microRNA research. Circ. Res. 2011, 108, 219–234.
  16. 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.
  17. Wang, H.; Meng, Q.; Qian, J.; Li, M.; Gu, C.; Yang, Y. Review: RNA-based diagnostic markers discovery and therapeutic targets development in cancer. Pharmacol. Ther. 2022, 234, 108123.
  18. Guo, Z.; Maki, M.; Ding, R.; Yang, Y.; Zhang, B.; Xiong, L. Genome-wide survey of tissue-specific microRNA and transcription factor regulatory networks in 12 tissues. Sci. Rep. 2014, 4, 5150.
  19. Wienholds, E.; Kloosterman, W.P.; Miska, E.; Alvarez-Saavedra, E.; Berezikov, E.; de Bruijn, E.; Horvitz, H.R.; Kauppinen, S.; Plasterk, R.H. MicroRNA expression in zebrafish embryonic development. Science 2005, 309, 310–311.
  20. Aboobaker, A.A.; Tomancak, P.; Patel, N.; Rubin, G.M.; Lai, E.C. Drosophila microRNAs exhibit diverse spatial expression patterns during embryonic development. Proc. Natl. Acad. Sci. USA 2005, 102, 18017–18022.
  21. Walker, J.C.; Harland, R.M. Expression of microRNAs during embryonic development of Xenopus tropicalis. Gene Expr. Patterns 2008, 8, 452–456.
  22. de Rie, D.; Abugessaisa, I.; Alam, T.; Arner, E.; Arner, P.; Ashoor, H.; Åström, G.; Babina, M.; Bertin, N.; Burroughs, A.M.; et al. An integrated expression atlas of miRNAs and their promoters in human and mouse. Nat. Biotechnol. 2017, 35, 872–878.
  23. Macconaill, L.E.; Garraway, L.A. Clinical implications of the cancer genome. J. Clin. Oncol. 2010, 28, 5219–5228.
  24. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46.
  25. Li, Y.; Kowdley, K.V. MicroRNAs in Common Human Diseases. Genom. Proteom. Bioinform. 2012, 10, 246–253.
  26. 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.
  27. Kozomara, A.; Birgaoanu, M.; Griffiths-Jones, S. miRBase: From microRNA sequences to function. Nucleic Acids Res. 2019, 47, D155–D162.
  28. Ludwig, N.; Leidinger, P.; Becker, K.; Backes, C.; Fehlmann, T.; Pallasch, C.; Rheinheimer, S.; Meder, B.; Stähler, C.; Meese, E.; et al. Distribution of miRNA expression across human tissues. Nucleic Acids Res. 2016, 44, 3865–3877.
  29. Beck, J.D.; Reidenbach, D.; Salomon, N.; Sahin, U.; Türeci, Ö.; Vormehr, M.; Kranz, L.M. mRNA therapeutics in cancer immunotherapy. Mol. Cancer 2021, 20, 69.
  30. De Guire, V.; Robitaille, R.; Tetreault, N.; Guerin, R.; Menard, C.; Bambace, N.; Sapieha, P. Circulating miRNAs as sensitive and specific biomarkers for the diagnosis and monitoring of human diseases: Promises and challenges. Clin. Biochem. 2013, 46, 846–860.
  31. Condrat, C.E.; Thompson, D.C.; Barbu, M.G.; Bugnar, O.L.; Boboc, A.; Cretoiu, D.; Suciu, N.; Cretoiu, S.M.; Voinea, S.C. miRNAs as Biomarkers in Disease: Latest Findings Regarding Their Role in Diagnosis and Prognosis. Cells 2020, 9, 276.
  32. Gareev, I.; de Jesus Encarnacion Ramirez, M.; Goncharov, E.; Ivliev, D.; Shumadalova, A.; Ilyasova, T.; Wang, C. MiRNAs and lncRNAs in the regulation of innate immune signaling. Noncoding RNA Res. 2023, 8, 534–541.
  33. Pauley, K.M.; Cha, S.; Chan, E.K. MicroRNA in autoimmunity and autoimmune diseases. J. Autoimmun. 2009, 32, 189–194.
  34. Sullivan, C.S.; Ganem, D. MicroRNAs and viral infection. Mol. Cell 2005, 20, 3–7.
  35. Skalsky, R.L.; Cullen, B.R. Viruses, microRNAs, and host interactions. Annu. Rev. Microbiol. 2010, 64, 123–141.
  36. Barbu, M.G.; Condrat, C.E.; Thompson, D.C.; Bugnar, O.L.; Cretoiu, D.; Toader, O.D.; Suciu, N.; Voinea, S.C. MicroRNA Involvement in Signaling Pathways During Viral Infection. Front. Cell Dev. Biol. 2020, 8, 143.
  37. Abu-Izneid, T.; AlHajri, N.; Ibrahim, A.M.; Javed, M.N.; Salem, K.M.; Pottoo, F.H.; Kamal, M.A. Micro-RNAs in the regulation of immune response against SARS CoV-2 and other viral infections. J. Adv. Res. 2021, 30, 133–145.
  38. Elfimova, N.; Schlattjan, M.; Sowa, J.P.; Dienes, H.P.; Canbay, A.; Odenthal, M. Circulating microRNAs: Promising candidates serving as novel biomarkers of acute hepatitis. Front. Physiol. 2012, 3, 476.
  39. Li, Y.J.; Xu, M.; Gao, Z.H.; Wang, Y.Q.; Yue, Z.; Zhang, Y.X.; Li, X.X.; Zhang, C.; Xie, S.Y.; Wang, P.Y. Alterations of serum levels of BDNF-related miRNAs in patients with depression. PLoS ONE 2013, 8, e63648.
  40. Scott, K.A.; Hoban, A.E.; Clarke, G.; Moloney, G.M.; Dinan, T.G.; Cryan, J.F. Thinking small: Towards microRNA-based therapeutics for anxiety disorders. Expert. Opin. Investig. Drugs 2015, 24, 529–542.
  41. Liu, W.; Liu, C.; Zhu, J.; Shu, P.; Yin, B.; Gong, Y.; Qiang, B.; Yuan, J.; Peng, X. MicroRNA-16 targets amyloid precursor protein to potentially modulate Alzheimer’s-associated pathogenesis in SAMP8 mice. Neurobiol. Aging 2012, 33, 522–534.
  42. Weir, D.W.; Sturrock, A.; Leavitt, B.R. Development of biomarkers for Huntington’s disease. Lancet Neurol. 2011, 10, 573–590.
  43. 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.
  44. Recchioni, R.; Marcheselli, F.; Olivieri, F.; Ricci, S.; Procopio, A.D.; Antonicelli, R. Conventional and novel diagnostic biomarkers of acute myocardial infarction: A promising role for circulating microRNAs. Biomarkers 2013, 18, 547–558.
  45. Nunez Lopez, Y.O.; Coen, P.M.; Goodpaster, B.H.; Seyhan, A.A. Gastric bypass surgery with exercise alters plasma microRNAs that predict improvements in cardiometabolic risk. Int. J. Obes. 2017, 41, 1121–1130.
  46. Nunez Lopez, Y.O.; Garufi, G.; Pasarica, M.; Seyhan, A.A. Elevated and Correlated Expressions of miR-24, miR-30d, miR-146a, and SFRP-4 in Human Abdominal Adipose Tissue Play a Role in Adiposity and Insulin Resistance. Int. J. Endocrinol. 2018, 2018, 7351902.
  47. Wang, Q.; Wang, Y.; Minto, A.W.; Wang, J.; Shi, Q.; Li, X.; Quigg, R.J. MicroRNA-377 is up-regulated and can lead to increased fibronectin production in diabetic nephropathy. FASEB J. 2008, 22, 4126–4135.
  48. Gambardella, S.; Rinaldi, F.; Lepore, S.M.; Viola, A.; Loro, E.; Angelini, C.; Vergani, L.; Novelli, G.; Botta, A. Overexpression of microRNA-206 in the skeletal muscle from myotonic dystrophy type 1 patients. J. Transl. Med. 2010, 8, 48.
  49. Seyhan, A.A.; Nunez Lopez, Y.O.; Xie, H.; Yi, F.; Mathews, C.; Pasarica, M.; Pratley, R.E. Pancreas-enriched miRNAs are altered in the circulation of subjects with diabetes: A pilot cross-sectional study. Sci. Rep. 2016, 6, 31479.
  50. Nunez Lopez, Y.O.; Garufi, G.; Seyhan, A.A. Altered levels of circulating cytokines and microRNAs in lean and obese individuals with prediabetes and type 2 diabetes. Mol. Biosyst. 2016, 13, 106–121.
  51. Nunez Lopez, Y.O.; Pittas, A.G.; Pratley, R.E.; Seyhan, A.A. Circulating levels of miR-7, miR-152 and miR-192 respond to vitamin D supplementation in adults with prediabetes and correlate with improvements in glycemic control. J. Nutr. Biochem. 2017, 49, 117–122.
  52. Raufi, A.G.; May, M.S.; Hadfield, M.J.; Seyhan, A.A.; El-Deiry, W.S. Advances in Liquid Biopsy Technology and Implications for Pancreatic Cancer. Int. J. Mol. Sci. 2023, 24, 4238.
  53. Smolarz, B.; Durczyński, A.; Romanowicz, H.; Hogendorf, P. The Role of microRNA in Pancreatic Cancer. Biomedicines 2021, 9, 1322.
  54. Kluiver, J.; Poppema, S.; de Jong, D.; Blokzijl, T.; Harms, G.; Jacobs, S.; Kroesen, B.J.; van den Berg, A. BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and diffuse large B cell lymphomas. J. Pathol. 2005, 207, 243–249.
  55. Lu, J.; Getz, G.; Miska, E.A.; Alvarez-Saavedra, E.; Lamb, J.; Peck, D.; Sweet-Cordero, A.; Ebert, B.L.; Mak, R.H.; Ferrando, A.A.; et al. MicroRNA expression profiles classify human cancers. Nature 2005, 435, 834–838.
  56. Ventura, A.; Jacks, T. MicroRNAs and cancer: Short RNAs go a long way. Cell 2009, 136, 586–591.
  57. Lawler, S.; Chiocca, E.A. Emerging functions of microRNAs in glioblastoma. J. Neurooncol. 2009, 92, 297–306.
  58. Suzuki, H.; Maruyama, R.; Yamamoto, E.; Kai, M. Epigenetic alteration and microRNA dysregulation in cancer. Front. Genet. 2013, 4, 258.
  59. Di Leva, G.; Garofalo, M.; Croce, C.M. MicroRNAs in cancer. Annu. Rev. Pathol. 2014, 9, 287–314.
  60. Hayes, J.; Peruzzi, P.P.; Lawler, S. MicroRNAs in cancer: Biomarkers, functions and therapy. Trends Mol. Med. 2014, 20, 460–469.
  61. Graveel, C.R.; Calderone, H.M.; Westerhuis, J.J.; Winn, M.E.; Sempere, L.F. Critical analysis of the potential for microRNA biomarkers in breast cancer management. Breast Cancer 2015, 7, 59–79.
  62. Lin, S.; Gregory, R.I. MicroRNA biogenesis pathways in cancer. Nat. Rev. Cancer 2015, 15, 321–333.
  63. Peng, Y.; Croce, C.M. The role of MicroRNAs in human cancer. Signal Transduct. Target. Ther. 2016, 1, 15004.
  64. Lulla, A.R.; Slifker, M.J.; Zhou, Y.; Lev, A.; Einarson, M.B.; Dicker, D.T.; El-Deiry, W.S. miR-6883 Family miRNAs Target CDK4/6 to Induce G(1) Phase Cell-Cycle Arrest in Colon Cancer Cells. Cancer Res. 2017, 77, 6902–6913.
  65. Biswas, S. MicroRNAs as Therapeutic Agents: The Future of the Battle against Cancer. Curr. Top. Med. Chem. 2018, 18, 2544–2554.
  66. Hu, W.; Tan, C.; He, Y.; Zhang, G.; Xu, Y.; Tang, J. Functional miRNAs in breast cancer drug resistance. Oncotargets Ther. 2018, 11, 1529–1541.
  67. Plotnikova, O.; Baranova, A.; Skoblov, M. Comprehensive Analysis of Human microRNA-mRNA Interactome. Front. Genet. 2019, 10, 933.
  68. Ali Syeda, Z.; Langden, S.S.S.; Munkhzul, C.; Lee, M.; Song, S.J. Regulatory Mechanism of MicroRNA Expression in Cancer. Int. J. Mol. Sci. 2020, 21, 1723.
  69. Annese, T.; Tamma, R.; De Giorgis, M.; Ribatti, D. microRNAs Biogenesis, Functions and Role in Tumor Angiogenesis. Front. Oncol. 2020, 10, 581007.
  70. Fathi, M.; Ghafouri-Fard, S.; Abak, A.; Taheri, M. Emerging roles of miRNAs in the development of pancreatic cancer. Biomed. Pharmacother. 2021, 141, 111914.
  71. Galka-Marciniak, P.; Urbanek-Trzeciak, M.O.; Nawrocka, P.M.; Kozlowski, P. A pan-cancer atlas of somatic mutations in miRNA biogenesis genes. Nucleic Acids Res. 2021, 49, 601–620.
  72. Pajares, M.J.; Alemany-Cosme, E.; Goni, S.; Bandres, E.; Palanca-Ballester, C.; Sandoval, J. Epigenetic Regulation of microRNAs in Cancer: Shortening the Distance from Bench to Bedside. Int. J. Mol. Sci. 2021, 22, 7350.
  73. Inoue, J.; Inazawa, J. Cancer-associated miRNAs and their therapeutic potential. J. Hum. Genet. 2021, 66, 937–945.
  74. Smolarz, B.; Durczyński, A.; Romanowicz, H.; Szyłło, K.; Hogendorf, P. miRNAs in Cancer (Review of Literature). Int. J. Mol. Sci. 2022, 23, 2805.
  75. Esquela-Kerscher, A.; Slack, F.J. Oncomirs—microRNAs with a role in cancer. Nat. Rev. Cancer 2006, 6, 259–269.
  76. Ding, J.; Cao, Y.; Qi, C.; Zong, Z. Dysregulated microRNAs participate in the crosstalk between colorectal cancer and atrial fibrillation. Hum. Cell 2023, 36, 1336–1342.
  77. Ricarte-Filho, J.C.; Casado-Medrano, V.; Reichenberger, E.; Spangler, Z.; Scheerer, M.; Isaza, A.; Baran, J.; Patel, T.; MacFarland, S.P.; Brodeur, G.M.; et al. DICER1 RNase IIIb domain mutations trigger widespread miRNA dysregulation and MAPK activation in pediatric thyroid cancer. Front. Endocrinol. 2023, 14, 1083382.
  78. Menon, A.; Abd-Aziz, N.; Khalid, K.; Poh, C.L.; Naidu, R. miRNA: A Promising Therapeutic Target in Cancer. Int. J. Mol. Sci. 2022, 23, 11502.
  79. Hata, A.; Lieberman, J. Dysregulation of microRNA biogenesis and gene silencing in cancer. Sci. Signal. 2015, 8, re3.
  80. Hata, A.; Kashima, R. Dysregulation of microRNA biogenesis machinery in cancer. Crit. Rev. Biochem. Mol. Biol. 2016, 51, 121–134.
  81. O’Carroll, D.; Schaefer, A. General principals of miRNA biogenesis and regulation in the brain. Neuropsychopharmacology 2013, 38, 39–54.
  82. Rodriguez, A.; Griffiths-Jones, S.; Ashurst, J.L.; Bradley, A. Identification of mammalian microRNA host genes and transcription units. Genome Res. 2004, 14, 1902–1910.
  83. Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233.
  84. Iwasaki, S.; Kobayashi, M.; Yoda, M.; Sakaguchi, Y.; Katsuma, S.; Suzuki, T.; Tomari, Y. Hsc70/Hsp90 chaperone machinery mediates ATP-dependent RISC loading of small RNA duplexes. Mol. Cell 2010, 39, 292–299.
  85. Shukla, G.C.; Singh, J.; Barik, S. MicroRNAs: Processing, Maturation, Target Recognition and Regulatory Functions. Mol. Cell Pharmacol. 2011, 3, 83–92.
  86. Akgul, B.; Erdogan, I. Intracytoplasmic Re-localization of miRISC Complexes. Front. Genet. 2018, 9, 403.
  87. Selbach, M.; Schwanhäusser, B.; Thierfelder, N.; Fang, Z.; Khanin, R.; Rajewsky, N. Widespread changes in protein synthesis induced by microRNAs. Nature 2008, 455, 58–63.
  88. Uhlmann, S.; Mannsperger, H.; Zhang, J.D.; Horvat, E.; Schmidt, C.; Küblbeck, M.; Henjes, F.; Ward, A.; Tschulena, U.; Zweig, K.; et al. Global microRNA level regulation of EGFR-driven cell-cycle protein network in breast cancer. Mol. Syst. Biol. 2012, 8, 570.
  89. Grimson, A.; Farh, K.K.; Johnston, W.K.; Garrett-Engele, P.; Lim, L.P.; Bartel, D.P. MicroRNA targeting specificity in mammals: Determinants beyond seed pairing. Mol. Cell 2007, 27, 91–105.
  90. Saetrom, P.; Heale, B.S.; Snøve, O., Jr.; Aagaard, L.; Alluin, J.; Rossi, J.J. Distance constraints between microRNA target sites dictate efficacy and cooperativity. Nucleic Acids Res. 2007, 35, 2333–2342.
  91. Desterro, J.; Bak-Gordon, P.; Carmo-Fonseca, M. Targeting mRNA processing as an anticancer strategy. Nat. Rev. Drug Discov. 2020, 19, 112–129.
  92. Vogelstein, B.; Papadopoulos, N.; Velculescu, V.E.; Zhou, S.; Diaz, L.A., Jr.; Kinzler, K.W. Cancer genome landscapes. Science 2013, 339, 1546–1558.
  93. Choudhury, Y.; Tay, F.C.; Lam, D.H.; Sandanaraj, E.; Tang, C.; Ang, B.T.; Wang, S. Attenuated adenosine-to-inosine editing of microRNA-376a* promotes invasiveness of glioblastoma cells. J. Clin. Investig. 2012, 122, 4059–4076.
  94. Stahlhut, C.; Slack, F.J. MicroRNAs and the cancer phenotype: Profiling, signatures and clinical implications. Genome Med. 2013, 5, 111.
  95. Dvinge, H.; Git, A.; Gräf, S.; Salmon-Divon, M.; Curtis, C.; Sottoriva, A.; Zhao, Y.; Hirst, M.; Armisen, J.; Miska, E.A.; et al. The shaping and functional consequences of the microRNA landscape in breast cancer. Nature 2013, 497, 378–382.
  96. Kim, T.M.; Huang, W.; Park, R.; Park, P.J.; Johnson, M.D. A developmental taxonomy of glioblastoma defined and maintained by MicroRNAs. Cancer Res. 2011, 71, 3387–3399.
  97. Manterola, L.; Guruceaga, E.; Gállego Pérez-Larraya, J.; González-Huarriz, M.; Jauregui, P.; Tejada, S.; Diez-Valle, R.; Segura, V.; Samprón, N.; Barrena, C.; et al. A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro Oncol. 2014, 16, 520–527.
  98. Kasinski, A.L.; Slack, F.J. MicroRNAs en route to the clinic: Progress in validating and targeting microRNAs for cancer therapy. Nat. Rev. Cancer 2011, 11, 849–864.
  99. Shankar, S.; Tien, J.C.-Y.; Siebenaler, R.F.; Chugh, S.; Dommeti, V.L.; Zelenka-Wang, S.; Wang, X.-M.; Apel, I.J.; Waninger, J.; Eyunni, S.; et al. An essential role for Argonaute 2 in EGFR-KRAS signaling in pancreatic cancer development. Nat. Commun. 2020, 11, 2817.
  100. Eder, M.; Scherr, M. MicroRNA and lung cancer. N. Engl. J. Med. 2005, 352, 2446–2448.
  101. Gonzales-Aloy, E.; Connerty, P.; Salik, B.; Liu, B.; Woo, A.J.; Haber, M.; Norris, M.D.; Wang, J.; Wang, J.Y. miR-101 suppresses the development of MLL-rearranged acute myeloid leukemia. Haematologica 2019, 104, e296–e299.
  102. Liu, Y.; Chen, X.; Cheng, R.; Yang, F.; Yu, M.; Wang, C.; Cui, S.; Hong, Y.; Liang, H.; Liu, M.; et al. The Jun/miR-22/HuR regulatory axis contributes to tumourigenesis in colorectal cancer. Mol. Cancer 2018, 17, 11.
  103. Zhu, S.; Cheng, X.; Wang, R.; Tan, Y.; Ge, M.; Li, D.; Xu, Q.; Sun, Y.; Zhao, C.; Chen, S.; et al. Restoration of microRNA function impairs MYC-dependent maintenance of MLL leukemia. Leukemia 2020, 34, 2484–2488.
  104. 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.
  105. Svoronos, A.A.; Engelman, D.M.; Slack, F.J. OncomiR or Tumor Suppressor? The Duplicity of MicroRNAs in Cancer. Cancer Res. 2016, 76, 3666–3670.
  106. Gambari, R.; Brognara, E.; Spandidos, D.A.; Fabbri, E. Targeting oncomiRNAs and mimicking tumor suppressor miRNAs: Νew trends in the development of miRNA therapeutic strategies in oncology (Review). Int. J. Oncol. 2016, 49, 5–32.
  107. Cho, W.C.S. OncomiRs: The discovery and progress of microRNAs in cancers. Mol. Cancer 2007, 6, 60.
  108. Frixa, T.; Donzelli, S.; Blandino, G. Oncogenic MicroRNAs: Key Players in Malignant Transformation. Cancers 2015, 7, 2466–2485.
  109. Zhou, K.; Liu, M.; Cao, Y. New Insight into microRNA Functions in Cancer: Oncogene-microRNA-Tumor Suppressor Gene Network. Front. Mol. Biosci. 2017, 4, 46.
  110. Shenouda, S.K.; Alahari, S.K. MicroRNA function in cancer: Oncogene or a tumor suppressor? Cancer Metastasis. Rev. 2009, 28, 369–378.
  111. Daoud, A.Z.; Mulholland, E.J.; Cole, G.; McCarthy, H.O. MicroRNAs in Pancreatic Cancer: Biomarkers, prognostic, and therapeutic modulators. BMC Cancer 2019, 19, 1130.
  112. Wang, H.; Peng, R.; Wang, J.; Qin, Z.; Xue, L. Circulating microRNAs as potential cancer biomarkers: The advantage and disadvantage. Clin. Epigenetics 2018, 10, 59.
  113. 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.
More
This entry is offline, you can click here to edit this entry!
Video Production Service