NcRNAs in Human Diseases: Comparison
Please note this is a comparison between Version 3 by Lily Guo and Version 2 by Eduardo Andrés-León.

The mammalian genome comprehends a small minority of genes that encode for proteins (barely 2% of the total genome in humans) and an immense majority of genes that are transcribed into RNA but not encoded for proteins (ncRNAs). These non-coding genes are intimately related to the expression regulation of protein-coding genes. The ncRNAs subtypes differ in their size, so there are long non-coding genes (lncRNAs) and other smaller ones, like microRNAs (miRNAs) and piwi-interacting RNAs (piRNAs).

Due to their important role in the maintenance of cellular functioning, any deregulation of the expression profiles of these ncRNAs can dissemble in the development of different types of diseases, such as cancer, neurodegenerative, or cardiovascular disorders.

  • ncRNAs
  • miRNAs
  • circRNAs
  • lncRNAs
  • piRNAs
  • diseases
  • cancer
  • cardiovascular disease
  • neurodegenerative
Please wait, diff process is still running!

References

  1. Melton, C.; Reuter, J.A.; Spacek, D.V.; Snyder, M. Recurrent somatic mutations in regulatory regions of human cancer genomes. Nat. Genet. 2015, 47, 710–716.
  2. Bartel, D.P. Metazoan MicroRNAs. Cell 2018, 173, 20–51.
  3. Batista, P.J.; Chang, H.Y. Long noncoding RNAs: Cellular address codes in development and disease. Cell 2013, 152, 1298–1307.
  4. Hanly, D.J.; Esteller, M.; Berdasco, M. Interplay between long non-coding RNAs and epigenetic machinery: Emerging targets in cancer? Philos. Trans. R. Soc. Lond. B Biol. Sci. 2018, 373, 20170074.
  5. Cech, T.R.; Steitz, J.A. The noncoding RNA revolution-trashing old rules to forge new ones. Cell 2014, 157, 77–94.
  6. Ishizu, H.; Siomi, H.; Siomi, M.C. Biology of PIWI-interacting RNAs: New insights into biogenesis and function inside and outside of germlines. Genes Dev. 2012, 26, 2361–2373.
  7. Sivagurunathan, S.; Arunachalam, J.P.; Chidambaram, S. PIWI-like protein, HIWI2 is aberrantly expressed in retinoblastoma cells and affects cell-cycle potentially through OTX2. Cell Mol. Biol. Lett. 2017, 22, 17.
  8. Karreth, F.A.; Tay, Y.; Perna, D.; Ala, U.; Tan, S.M.; Rust, A.G.; DeNicola, G.; Webster, K.A.; Weiss, D.; Perez-Mancera, P.A.; et al. In Vivo Identification of Tumor-Suppressive PTEN ceRNAs in an Oncogenic BRAF-Induced Mouse Model of Melanoma. Cell 2011, 147, 382–395.
  9. Song, C.; Zhang, J.; Qi, H.P.; Feng, C.C.; Chen, Y.P.; Cao, Y.G.; Ba, L.N.; Ai, B.; Wang, Q.Y.; Huang, W.; et al. The global view of mRNA-related ceRNA cross-talks across cardiovascular diseases. Sci. Rep. 2017, 7.
  10. Ghoussaini, M.; Mountjoy, E.; Carmona, M.; Peat, G.; Schmidt, E.M.; Hercules, A.; Fumis, L.; Miranda, A.; Carvalho-Silva, D.; Buniello, A.; et al. Open Targets Genetics: Systematic identification of trait-associated genes using large-scale genetics and functional genomics. Nucleic Acids Res. 2021, 49, D1311–D1320.
  11. Gong, J.; Tong, Y.; Zhang, H.M.; Wang, K.; Hu, T.; Shan, G.; Sun, J.; Guo, A.Y. Genome-Wide Identification of SNPs in MicroRNA Genes and the SNP Effects on MicroRNA Target Binding and Biogenesis. Hum. Mutat. 2012, 33, 254–263.
  12. Tanic, M.; Andres, E.; Rodriguez-Pinilla, S.M.; Marquez-Rodas, I.; Cebollero-Presmanes, M.; Fernandez, V.; Osorio, A.; Benitez, J.; Martinez-Delgado, B. MicroRNA-based molecular classification of non-BRCA1/2 hereditary breast tumours. Br. J. Cancer 2013, 109, 2724–2734.
  13. Freedman, M.L.; Monteiro, A.N.; Gayther, S.A.; Coetzee, G.A.; Risch, A.; Plass, C.; Casey, G.; De Biasi, M.; Carlson, C.; Duggan, D.; et al. Principles for the post-GWAS functional characterization of cancer risk loci. Nat. Genet. 2011, 43, 513–518.
  14. Andres-Leon, E.; Cases, I.; Alonso, S.; Rojas, A.M. Novel miRNA-mRNA interactions conserved in essential cancer pathways. Sci. Rep. 2017, 7, 46101.
  15. Esquela-Kerscher, A.; Slack, F.J. Oncomirs—microRNAs with a role in cancer. Nat. Rev. Cancer 2006, 6, 259–269.
  16. Hammond, S.M. MicroRNAs as oncogenes. Curr. Opin. Genet. Dev. 2006, 16, 4–9.
  17. Hossain, A.; Kuo, M.T.; Saunders, G.F. Mir-17-5p regulates breast cancer cell proliferation by inhibiting translation of AIB1 mRNA. Mol. Cell Biol. 2006, 26, 8191–8201.
  18. Akao, Y.; Nakagawa, Y.; Naoe, T. MicroRNAs 143 and 145 are possible common onco-microRNAs in human cancers. Oncol. Rep. 2006, 16, 845–850.
  19. Suzuki, H.I.; Yamagata, K.; Sugimoto, K.; Iwamoto, T.; Kato, S.; Miyazono, K. Modulation of microRNA processing by p53. Nature 2009, 460, 529–533.
  20. Kent, O.A.; Chivukula, R.R.; Mullendore, M.; Wentzel, E.A.; Feldmann, G.; Lee, K.H.; Liu, S.; Leach, S.D.; Maitra, A.; Mendell, J.T. Repression of the miR-143/145 cluster by oncogenic Ras initiates a tumor-promoting feed-forward pathway. Genes Dev. 2010, 24, 2754–2759.
  21. Yuan, F.; Sun, R.F.; Li, L.J.; Jin, B.; Wang, Y.Y.; Liang, Y.D.; Che, G.L.; Gao, L.B.; Zhang, L. A functional variant rs353292 in the flanking region of miR-143/145 contributes to the risk of colorectal cancer. Sci. Rep. 2016, 6.
  22. Janaszak-Jasiecka, A.; Bartoszewska, S.; Kochan, K.; Piotrowski, A.; Kalinowski, L.; Kamysz, W.; Ochocka, R.J.; Bartoszewski, R.; Collawn, J.F. miR-429 regulates the transition between Hypoxia-Inducible Factor (HIF)1A and HIF3A expression in human endothelial cells. Sci. Rep. 2016, 6, 22775.
  23. Lu, Y.; Zhao, X.; Liu, Q.; Li, C.; Graves-Deal, R.; Cao, Z.; Singh, B.; Franklin, J.L.; Wang, J.; Hu, H.; et al. lncRNA MIR100HG-derived miR-100 and miR-125b mediate cetuximab resistance via Wnt/beta-catenin signaling. Nat. Med. 2017, 23, 1331–1341.
  24. Sevignani, C.; Calin, G.A.; Nnadi, S.C.; Shimizu, M.; Davuluri, R.V.; Hyslop, T.; Demant, P.; Croce, C.M.; Siracusa, L.D. MicroRNA genes are frequently located near mouse cancer susceptibility loci. Proc. Natl. Acad. Sci. USA 2007, 104, 8017–8022.
  25. Suzuki, H.; Maruyama, R.; Yamamoto, E.; Kai, M. Epigenetic alteration and microRNA dysregulation in cancer. Front. Genet. 2013, 4, 258.
  26. O’Donnell, K.A.; Wentzel, E.A.; Zeller, K.I.; Dang, C.V.; Mendell, J.T. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 2005, 435, 839–843.
  27. Viswanathan, S.R.; Powers, J.T.; Einhorn, W.; Hoshida, Y.; Ng, T.L.; Toffanin, S.; O’Sullivan, M.; Lu, J.; Phillips, L.A.; Lockhart, V.L.; et al. Lin28 promotes transformation and is associated with advanced human malignancies. Nat. Genet. 2009, 41, 843–848.
  28. Yang, Q.; Jie, Z.; Ye, S.; Li, Z.; Han, Z.; Wu, J.; Yang, C.; Jiang, Y. Genetic variations in miR-27a gene decrease mature miR-27a level and reduce gastric cancer susceptibility. Oncogene 2014, 33, 193–202.
  29. Hon, C.C.; Ramilowski, J.A.; Harshbarger, J.; Bertin, N.; Rackham, O.J.L.; Gough, J.; Denisenko, E.; Schmeier, S.; Poulsen, T.M.; Severin, J.; et al. An atlas of human long non-coding RNAs with accurate 5 ‘ ends. Nature 2017, 543, 199–204.
  30. Zhu, J.; Liu, S.; Ye, F.; Shen, Y.; Tie, Y.; Zhu, J.; Wei, L.; Jin, Y.; Fu, H.; Wu, Y.; et al. Long Noncoding RNA MEG3 Interacts with p53 Protein and Regulates Partial p53 Target Genes in Hepatoma Cells. PLoS ONE 2015, 10, e0139790.
  31. Zhang, J.; Yao, T.; Wang, Y.; Yu, J.; Liu, Y.; Lin, Z. Long noncoding RNA MEG3 is downregulated in cervical cancer and affects cell proliferation and apoptosis by regulating miR-21. Cancer Biol. Ther. 2016, 17, 104–113.
  32. Ying, L.; Huang, Y.; Chen, H.; Wang, Y.; Xia, L.; Chen, Y.; Liu, Y.; Qiu, F. Downregulated MEG3 activates autophagy and increases cell proliferation in bladder cancer. Mol. Biosyst. 2013, 9, 407–411.
  33. Conte, F.; Fiscon, G.; Chiara, M.; Colombo, T.; Farina, L.; Paci, P. Role of the long non-coding RNA PVT1 in the dysregulation of the ceRNA-ceRNA network in human breast cancer. PLoS ONE 2017, 12, e0171661.
  34. Perry, M.M.; Tsitsiou, E.; Austin, P.J.; Lindsay, M.A.; Gibeon, D.S.; Adcock, I.M.; Chung, K.F. Role of non-coding RNAs in maintaining primary airway smooth muscle cells. Respir. Res. 2014, 15, 58.
  35. Zhang, L.R.; Lin, J.; Ye, Y.Q.; Oba, T.; Gentile, E.; Lian, J.; Wang, J.; Zhao, Y.; Gu, J.; Wistuba, I.I.; et al. Serum MicroRNA-150 Predicts Prognosis for Early-Stage Non-Small Cell Lung Cancer and Promotes Tumor Cell Proliferation by Targeting Tumor Suppressor Gene SRCIN1. Clin. Pharmacol. Ther. 2018, 103, 1061–1073.
  36. Ghisi, M.; Corradin, A.; Basso, K.; Frasson, C.; Serafin, V.; Mukherjee, S.; Mussolin, L.; Ruggero, K.; Bonanno, L.; Guffanti, A.; et al. Modulation of microRNA expression in human T-cell development: Targeting of NOTCH3 by miR-150. Blood 2011, 117, 7053–7062.
  37. George, J.; Lim, J.S.; Jang, S.J.; Cun, Y.P.; Ozretic, L.; Kong, G.; Leenders, F.; Lu, X.; Fernandez-Cuesta, L.; Bosco, G.; et al. Comprehensive genomic profiles of small cell lung cancer. Nature 2015, 524, 47–53.
  38. Sun, Q.; Wang, Y.Z.; Fan, J.Y.; Li, Z.H.; Zhang, J.H.; Wang, L.J.; Fan, X.K.; Ji, M.M.; Zhu, M.; Dai, J.C.; et al. Association of expression quantitative trait loci for long noncoding RNAs with lung cancer risk in Asians. Mol. Carcinog. 2019, 58, 1303–1313.
  39. Siddiqi, S.; Matushansky, I. Piwis and piwi-interacting RNAs in the epigenetics of cancer. J. Cell. Biochem. 2012, 113, 373–380.
  40. Fu, A.; Jacobs, D.I.; Zhu, Y. Epigenome-wide analysis of piRNAs in gene-specific DNA methylation. RNA Biol. 2014, 11, 1301–1312.
  41. Huang, G.; Hu, H.; Xue, X.; Shen, S.; Gao, E.; Guo, G.; Shen, X.; Zhang, X. Altered expression of piRNAs and their relation with clinicopathologic features of breast cancer. Clin. Transl. Oncol. 2013, 15, 563–568.
  42. Jacobs, D.I.; Qin, Q.; Lerro, M.C.; Fu, A.; Dubrow, R.; Claus, E.B.; DeWan, A.T.; Wang, G.L.; Lin, H.F.; Zhu, Y. PIWI-Interacting RNAs in Gliomagenesis: Evidence from Post-GWAS and Functional Analyses. Cancer Epidemiol. Biomark. Prev. 2016, 25, 1073–1080.
  43. Rupaimoole, R.; Calin, G.A.; Lopez-Berestein, G.; Sood, A.K. miRNA Deregulation in Cancer Cells and the Tumor Microenvironment. Cancer Discov. 2016, 6, 235–246.
  44. Guo, J.U.; Agarwal, V.; Guo, H.; Bartel, D.P. Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 2014, 15, 409.
  45. Salzman, J.; Chen, R.E.; Olsen, M.N.; Wang, P.L.; Brown, P.O. Cell-type specific features of circular RNA expression. PLoS Genet. 2013, 9, e1003777.
  46. Bachmayr-Heyda, A.; Reiner, A.T.; Auer, K.; Sukhbaatar, N.; Aust, S.; Bachleitner-Hofmann, T.; Mesteri, I.; Grunt, T.W.; Zeillinger, R.; Pils, D. Correlation of circular RNA abundance with proliferation--exemplified with colorectal and ovarian cancer, idiopathic lung fibrosis, and normal human tissues. Sci. Rep. 2015, 5, 8057.
  47. Kadakkuzha, B.M.; Liu, X.A.; McCrate, J.; Shankar, G.; Rizzo, V.; Afinogenova, A.; Young, B.; Fallahi, M.; Carvalloza, A.C.; Raveendra, B.; et al. Transcriptome analyses of adult mouse brain reveal enrichment of lncRNAs in specific brain regions and neuronal populations. Front. Cell. Neurosci. 2015, 9, 63.
  48. Rybak-Wolf, A.; Stottmeister, C.; Glazar, P.; Jens, M.; Pino, N.; Giusti, S.; Hanan, M.; Behm, M.; Bartok, O.; Ashwal-Fluss, R.; et al. Circular RNAs in the Mammalian Brain Are Highly Abundant, Conserved, and Dynamically Expressed. Mol. Cell 2015, 58, 870–885.
  49. You, X.; Vlatkovic, I.; Babic, A.; Will, T.; Epstein, I.; Tushev, G.; Akbalik, G.; Wang, M.; Glock, C.; Quedenau, C.; et al. Neural circular RNAs are derived from synaptic genes and regulated by development and plasticity. Nat. Neurosci. 2015, 18, 603–610.
  50. Davis, G.M.; Haas, M.A.; Pocock, R. MicroRNAs: Not “Fine-Tuners” but Key Regulators of Neuronal Development and Function. Front. Neurol. 2015, 6, 245.
  51. Ng, S.Y.; Johnson, R.; Stanton, L.W. Human long non-coding RNAs promote pluripotency and neuronal differentiation by association with chromatin modifiers and transcription factors. EMBO J. 2012, 31, 522–533.
  52. Schratt, G. microRNAs at the synapse. Nat. Rev. Neurosci. 2009, 10, 842–849.
  53. Junn, E.; Lee, K.W.; Jeong, B.S.; Chan, T.W.; Im, J.Y.; Mouradian, M.M. Repression of alpha-synuclein expression and toxicity by microRNA-7. Proc. Natl. Acad. Sci. USA 2009, 106, 13052–13057.
  54. Bak, M.; Silahtaroglu, A.; Moller, M.; Christensen, M.; Rath, M.F.; Skryabin, B.; Tommerup, N.; Kauppinen, S. MicroRNA expression in the adult mouse central nervous system. RNA 2008, 14, 432–444.
  55. Ashwal-Fluss, R.; Meyer, M.; Pamudurti, N.R.; Ivanov, A.; Bartok, O.; Hanan, M.; Evantal, N.; Memczak, S.; Rajewsky, N.; Kadener, S. circRNA biogenesis competes with pre-mRNA splicing. Mol. Cell 2014, 56, 55–66.
  56. Elstner, M.; Morris, C.M.; Heim, K.; Lichtner, P.; Bender, A.; Mehta, D.; Schulte, C.; Sharma, M.; Hudson, G.; Goldwurm, S.; et al. Single-Cell Expression Profiling of Dopaminergic Neurons Combined with Association Analysis Identifies Pyridoxal Kinase as Parkinson’s Disease Gene. Ann. Neurol. 2009, 66, 792–798.
  57. Giraldez, T.; Dominguez, J.; Alvarez de la Rosa, D. ENaC in the brain--future perspectives and pharmacological implications. Curr. Mol. Pharmacol. 2013, 6, 44–49.
  58. Mortezaei, Z.; Lanjanian, H.; Masoudi-Nejad, A. Candidate novel long noncoding RNAs, MicroRNAs and putative drugs for Parkinson’s disease using a robust and efficient genome-wide association study. Genomics 2017, 109, 158–164.
  59. Liu, Y.; Chen, Y.Y.; Liu, H.; Yao, C.J.; Zhu, X.X.; Chen, D.J.; Yang, J.; Lu, Y.J.; Cao, J.Y. Association between ubiquitin carboxy-terminal hydrolase-L1 S18Y variant and risk of Parkinson’s disease: The impact of ethnicity and onset age. Neurol. Sci. 2015, 36, 179–188.
  60. Carrieri, C.; Cimatti, L.; Biagioli, M.; Beugnet, A.; Zucchelli, S.; Fedele, S.; Pesce, E.; Ferrer, I.; Collavin, L.; Santoro, C.; et al. Long non-coding antisense RNA controls Uchl1 translation through an embedded SINEB2 repeat. Nature 2012, 491, 454–457.
  61. Lukiw, W.J. Circular RNA (circRNA) in Alzheimer’s disease (AD). Front. Genet. 2013, 4, 307.
  62. Zhang, S.; Zhu, D.; Li, H.; Li, H.; Feng, C.; Zhang, W. Characterization of circRNA-Associated-ceRNA Networks in a Senescence-Accelerated Mouse Prone 8 Brain. Mol. Ther. 2017, 25, 2053–2061.
  63. Wei, C.W.; Luo, T.; Zou, S.S.; Wu, A.S. The Role of Long Noncoding RNAs in Central Nervous System and Neurodegenerative Diseases. Front. Behav. Neurosci. 2018, 12, 175.
  64. Massone, S.; Vassallo, I.; Fiorino, G.; Castelnuovo, M.; Barbieri, F.; Borghi, R.; Tabaton, M.; Robello, M.; Gatta, E.; Russo, C.; et al. 17A, a novel non-coding RNA, regulates GABA B alternative splicing and signaling in response to inflammatory stimuli and in Alzheimer disease. Neurobiol. Dis. 2011, 41, 308–317.
  65. Hebert, S.S.; De Strooper, B. Alterations of the microRNA network cause neurodegenerative disease. Trends Neurosci. 2009, 32, 199–206.
  66. Vilardo, E.; Barbato, C.; Ciotti, M.; Cogoni, C.; Ruberti, F. MicroRNA-101 regulates amyloid precursor protein expression in hippocampal neurons. J. Biol. Chem. 2010, 285, 18344–18351.
  67. Han, Z.J.; Xue, W.W.; Tao, L.; Zhu, F. Identification of Key Long Non-Coding RNAs in the Pathology of Alzheimer’s Disease and their Functions Based on Genome-Wide Associations Study, Microarray, and RNA-seq Data. J. Alzheimers Dis. 2019, 68, 339–355.
  68. Kataoka, M.; Huang, Z.P.; Wang, D.Z. Build a braveheart: The missing linc (RNA). Circ. Res. 2013, 112, 1532–1534.
  69. Hou, J.; Long, H.; Zhou, C.; Zheng, S.; Wu, H.; Guo, T.; Wu, Q.; Zhong, T.; Wang, T. Long noncoding RNA Braveheart promotes cardiogenic differentiation of mesenchymal stem cells in vitro. Stem Cell Res. Ther. 2017, 8, 4.
  70. Viereck, J.; Kumarswamy, R.; Foinquinos, A.; Xiao, K.; Avramopoulos, P.; Kunz, M.; Dittrich, M.; Maetzig, T.; Zimmer, K.; Remke, J.; et al. Long noncoding RNA Chast promotes cardiac remodeling. Sci. Transl. Med. 2016, 8, 326ra322.
  71. Stępień, E.; Costa, M.C.; Kurc, S.; Drozdz, A.; Cortez-Dias, N.; Enguita, F.J. The circulating non-coding RNA landscape for biomarker research: Lessons and prospects from cardiovascular diseases. Acta Pharmacol. Sin. 2018, 39, 1085–1099.
  72. Jarinova, O.; Stewart, A.F.R.; Roberts, R.; Wells, G.; Lau, P.; Naing, T.; Buerki, C.; McLean, B.W.; Cook, R.C.; Parker, J.S.; et al. Functional Analysis of the Chromosome 9p21.3 Coronary Artery Disease Risk Locus. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 1671–U1670.
  73. Arslan, S.; Berkan, O.; Lalem, T.; Ozbilum, N.; Goksel, S.; Korkmaz, O.; Cetin, N.; Devaux, Y.; Cardiolinc, N. Long non-coding RNAs in the atherosclerotic plaque. Atherosclerosis 2017, 266, 176–181.
  74. Nikpay, M.; Beehler, K.; Valsesia, A.; Hager, J.; Harper, M.E.; Dent, R.; McPherson, R. Genome-wide identification of circulating-miRNA expression quantitative trait loci reveals the role of severalmiRNAs in the regulation of cardiometabolic phenotypes. Cardiovasc. Res. 2019, 115, 1629–1645.
  75. Li, Y.; Qiu, C.; Tu, J.; Geng, B.; Yang, J.; Jiang, T.; Cui, Q. HMDD v2.0: A database for experimentally supported human microRNA and disease associations. Nucleic Acids Res. 2014, 42, D1070–D1074.
  76. Xu, S.; Kamato, D.; Little, P.J.; Nakagawa, S.; Pelisek, J.; Jin, Z.G. Targeting epigenetics and non-coding RNAs in atherosclerosis: from mechanisms to therapeutics. Pharmacol. Ther. 2019, 196, 15–43.
  77. Eken, S.M.; Jin, H.; Chernogubova, E.; Li, Y.; Simon, N.; Sun, C.; Korzunowicz, G.; Busch, A.; Backlund, A.; Osterholm, C.; et al. MicroRNA-210 Enhances Fibrous Cap Stability in Advanced Atherosclerotic Lesions. Circ. Res. 2017, 120, 633–644.
  78. Jin, H.; Li, D.Y.; Chernogubova, E.; Sun, C.; Busch, A.; Eken, S.M.; Saliba-Gustafsson, P.; Winter, H.; Winski, G.; Raaz, U.; et al. Local Delivery of miR-21 Stabilizes Fibrous Caps in Vulnerable Atherosclerotic Lesions. Mol. Ther. 2018, 26, 1040–1055.
  79. Bye, A.; Rosjo, H.; Nauman, J.; Silva, G.J.; Follestad, T.; Omland, T.; Wisloff, U. Circulating microRNAs predict future fatal myocardial infarction in healthy individuals—The HUNT study. J. Mol. Cell. Cardiol. 2016, 97, 162–168.
  80. Matsumoto, S.; Sakata, Y.; Suna, S.; Nakatani, D.; Usami, M.; Hara, M.; Kitamura, T.; Hamasaki, T.; Nanto, S.; Kawahara, Y.; et al. Circulating p53-responsive microRNAs are predictive indicators of heart failure after acute myocardial infarction. Circ. Res. 2013, 113, 322–326.
  81. Zhang, X.O.; Dong, R.; Zhang, Y.; Zhang, J.L.; Luo, Z.; Zhang, J.; Chen, L.L.; Yang, L. Diverse alternative back-splicing and alternative splicing landscape of circular RNAs. Genome Res. 2016, 26, 1277–1287.
  82. Yan, Y.; Zhang, B.; Liu, N.; Qi, C.; Xiao, Y.; Tian, X.; Li, T.; Liu, B. Circulating Long Noncoding RNA UCA1 as a Novel Biomarker of Acute Myocardial Infarction. Biomed. Res. Int. 2016, 2016, 8079372.
  83. Du, W.W.; Zhang, C.; Yang, W.N.; Yong, T.Q.; Awan, F.M.; Yang, B.B. Identifying and Characterizing circRNA-Protein Interaction. Theranostics 2017, 7, 4183–4191.
  84. Grieco, F.A.; Schiavo, A.A.; Brozzi, F.; Juan-Mateu, J.; Bugliani, M.; Marchetti, P.; Eizirik, D. The microRNAs miR-211-5p and miR-204-5p modulate ER stress in human beta cells. J. Mol. Endocrinol. 2019, 63, 139–149.
  85. Zhao, X.; Mohan, R.; Ozcan, S.; Tang, X. MicroRNA-30d induces insulin transcription factor MafA and insulin production by targeting mitogen-activated protein 4 kinase 4 (MAP4K4) in pancreatic beta-cells. J. Biol. Chem. 2012, 287, 31155–31164.
  86. El Ouaamari, A.; Baroukh, N.; Martens, G.A.; Lebrun, P.; Pipeleers, D.; van Obberghen, E. miR-375 targets 3′-phosphoinositide-dependent protein kinase-1 and regulates glucose-induced biological responses in pancreatic beta-cells. Diabetes 2008, 57, 2708–2717.
  87. Hu, S.; Zhang, M.; Sun, F.; Ren, L.; He, X.; Hua, J.; Peng, S. miR-375 controls porcine pancreatic stem cell fate by targeting 3-phosphoinositide-dependent protein kinase-1 (Pdk1). Cell. Prolif. 2016, 49, 395–406.
  88. Poy, M.N.; Eliasson, L.; Krutzfeldt, J.; Kuwajima, S.; Ma, X.; Macdonald, P.E.; Pfeffer, S.; Tuschl, T.; Rajewsky, N.; Rorsman, P.; et al. A pancreatic islet-specific microRNA regulates insulin secretion. Nature 2004, 432, 226–230.
  89. Zhou, Y.; Gu, P.; Shi, W.; Li, J.; Hao, Q.; Cao, X.; Lu, Q.; Zeng, Y. MicroRNA-29a induces insulin resistance by targeting PPARdelta in skeletal muscle cells. Int. J. Mol. Med. 2016, 37, 931–938.
  90. Yang, W.M.; Jeong, H.J.; Park, S.Y.; Lee, W. Induction of miR-29a by saturated fatty acids impairs insulin signaling and glucose uptake through translational repression of IRS-1 in myocytes. FEBS Lett. 2014, 588, 2170–2176.
  91. Patel, V.; Hajarnis, S.; Williams, D.; Hunter, R.; Huynh, D.; Igarashi, P. MicroRNAs regulate renal tubule maturation through modulation of Pkd1. J. Am. Soc. Nephrol. 2012, 23, 1941–1948.
  92. Wang, G.; Kwan, B.C.; Lai, F.M.; Chow, K.M.; Li, P.K.; Szeto, C.C. Elevated levels of miR-146a and miR-155 in kidney biopsy and urine from patients with IgA nephropathy. Dis. Markers 2011, 30, 171–179.
  93. Luo, X.B.; Yang, W.L.; Ye, D.Q.; Cui, H.J.; Zhang, Y.; Hirankarn, N.; Qian, X.X.; Tang, Y.J.; Lau, Y.L.; de Vries, N.; et al. A Functional Variant in MicroRNA-146a Promoter Modulates Its Expression and Confers Disease Risk for Systemic Lupus Erythematosus. Plos Genet. 2011, 7.
  94. Xue, Z.X.; Cui, C.J.; Liao, Z.J.; Xia, S.W.; Zhang, P.J.; Qin, J.L.; Guo, Q.; Chen, S.; Fu, Q.; Yin, Z.H.; et al. Identification of LncRNA Linc00513 Containing Lupus-Associated Genetic Variants as a Novel Regulator of Interferon Signaling Pathway. Front. Immunol. 2018, 9.
More
Video Production Service