Русская версия English version   
Том 16   Выпуск 2   Год 2021
Назипова Н.Н.

Разнообразие некодирующих РНК в геномах эукариот

Математическая биология и биоинформатика. 2021;16(2):256-298.

doi: 10.17537/2021.16.256.

Список литературы

  1. International Human Genome Sequencing Consortium. Finishing the euchromatic sequence of the human genome. Nature. 2004;431:931–945. doi: 10.1038/nature03001
  2. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74. doi: 10.1038/nature11247
  3. Clark M.B., Amaral P.P., Schlesinger F.J., Dinger M.E., Taft R.J., Rinn J.L., Ponting C.P., Stadler P.F., Morris K.V., Morillon A. et al. The reality of pervasive transcription. PLoS Biol. 2011;9:e1000625. doi: 10.1371/journal.pbio.1000625
  4. Carninci P., Kasukawa T., Katayama S., Gough J., Frith M.C., Maeda N., Oyama R., Ravasi T., Lenhard B., Wells C. et al. The transcriptional landscape of the mammalian genome. Science. 2005;309:1559–1563. doi: 10.1126/science.1112014
  5. Katayama S., Tomaru Y., Kasukawa T., Waki K., Nakanishi M., Nakamura M., Nishida H., Yap C.C., Suzuki M., Kawai J. et al. Antisense transcription in the mammalian transcriptome. Science. 2005;309:1564–1566. doi: 10.1126/science.1112009
  6. Birney E., Stamatoyannopoulos J.A., Dutta A., Guigo R., Gingeras T.R., Margulies E.H., Weng Z., Snyder M., Dermitzakis E.T., Thurman R.E. et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 2007;447:799–816. doi: 10.1038/nature05874
  7. Kapranov P., Cheng J., Dike S., Nix D.A., Duttagupta R., Willingham A.T., Stadler P.F., Hertel J., Hackermuller J., Hofacker I.L. et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science. 2007;316:1484–1488. doi: 10.1126/science.1138341
  8. Bertone P., Stolc V., Royce T.E., Rozowsky J.S., Urban A.E., Zhu X., Rinn J.L., Tongprasit W., Samanta M., Weissman S. et al. Global identification of human transcribed sequences with genome tiling arrays. Science. 2004;306:2242–2246. doi: 10.1126/science.1103388
  9. Cheng J., Kapranov P., Drenkow J., Dike S., Brubaker S., Patel S., Long J., Stern D., Tammana H., Helt G. et al. Transcriptional maps of 10 human chromosomes at 5-nucleotide resolution. Science. 2005;308:1149–1154. doi: 10.1126/science.1108625
  10. Kapranov P., Cawley S.E., Drenkow J., Bekiranov S., Strausberg R.L., Fodor S.P.A., Gingeras T.R. Large-scale transcriptional activity in chromosomes 21 and 22. Science. 2002;296:916–919. doi: 10.1126/science.1068597
  11. Rinn J.L., Euskirchen G., Bertone P., Martone R., Luscombe N.M., Hartman S., Harrison P.M., Nelson F.K., Miller P., Gerstein M. et al. The transcriptional activity of human Chromosome 22. Genes Dev. 2003;17:529–540. doi: 10.1101/gad.1055203
  12. Kapranov P., Willingham A.T., Gingeras T.R. Genome-wide transcription and the implications for genomic organization. Nat. Rev. Genet. 2007;8:413–423. doi: 10.1038/nrg2083
  13. Wang J., Zhang J., Zheng H., Li J., Liu D., Li H., Samudrala R., Yu J., Wong G.K. Mouse transcriptome: Neutral evolution of ‘non-coding’ complementary DNAs. Nature. 2004;431:757. doi: 10.1038/nature03016
  14. Struhl K. Transcriptional noise and the fidelity of initiation by RNA polymerase II. Nat. Struct. Mol. Biol. 2007;14:103–105. doi: 10.1038/nsmb0207-103
  15. Ebisuya M., Yamamoto T., Nakajima M., Nishida E. Ripples from neighbouring transcription. Nat. Cell Biol. 2008;10:1106–1113. doi: 10.1038/ncb1771
  16. Palazzo A.F., Lee E.S. Non-coding RNA: what is functional and what is junk? Front. Genet. 2015;6. doi: 10.3389/fgene.2015.00002
  17. Mattick J.S., Taft R.J., Faulkner G.J. A global view of genomic information–moving beyond the gene and the master regulator. Trends Genet. 2010;26:21–28 doi: 10.1016/j.tig.2009.11.002
  18. Huttenhofer A., Brosius J., Bachellerie J.P. RNomics: identification and function of small, non-messenger RNAs. Curr. Opin. Chem. Biol. 2002;6:835–843. doi: 10.1016/S1367-5931(02)00397-6
  19. Huttenhofer A., Schattner P., Polacek N. Non-coding RNAs: hope or hype? Trends Genet. 2005;21:289–297. doi: 10.1016/j.tig.2005.03.007
  20. Eddy S.R. Non-coding RNA genes and the modern RNA world. Nature Reviews Genetics. 2001;2:919–929. doi: 10.1038/35103511
  21. Kawano M., Reynolds A.A., Miranda-Rios J., Storz G. Detection of 5′ and 3′-UTR-derived small RNAs and cis-encoded antisense RNAs in Escherichia coli. Nucleic Acids Res. 2005;33:1040–1050. doi: 10.1093/nar/gki256
  22. Mattick J.S. RNA regulation: a new genetics? Nat. Rev. Genet. 2004;5:316–323. doi: 10.1038/nrg1321
  23. Mattick J.S., Makunin I.V. Small regulatory RNAs in mammals. Hum. Mol. Genet. 2005;14. P. R121–R132. doi: 10.1093/hmg/ddi101
  24. Weinberg R.A., Penman S. Small molecular weight monodisperse nuclear RNA. J. Mol. Biol. 1968;38:289–304. doi: 10.1016/0022-2836(68)90387-2
  25. Zieve G.W. Two groups of small stable RNAs. Cell. 1981;25:296–297. doi: 10.1016/0092-8674(81)90046-5
  26. Busch H., Reddy R., Rothblum L., Choi Y.C. SnRNAs, SnRNPs, and RNA processing. Annu. Rev. Biochem. 1982;51:617–654. doi: 10.1146/annurev.bi.51.070182.003153
  27. Yang V.W., Lerner M.R., Steitz J.A., Flint S.J. A small nuclear ribonucleoprotein is required for splicing of adenoviral early RNA sequences. PNAS. 1981;78:1371–1375. doi: 10.1073/pnas.78.3.1371
  28. Yu Y.T., Scharl E.C., Smith C.M., Steitz J.A. In: The RNA World, 2nd edn. Eds. Gesteland R.F., Cech T.R., Atkins J.F. New York: Cold Spring Harbor Laboratory Press, 1999. P. 487–524.
  29. Burge C.B., Tuschl T., Sharp P.A. Splicing of precursors to mRNAs by the spliceosome. In: The RNA World, 2nd edn. Eds. Gesteland R.F., Cech T.R., Atkins J.F. New York: Cold Spring Harbor Laboratory Press, 1999. P. 525–560.
  30. Tarn W.Y., Steitz J.A. Highly diverged U4 and U6 small nuclear RNAs required for splicing rare AT-AC introns. Science. 1996;273:1824–1832. doi: 10.1126/science.273.5283.1824
  31. Sharp P.A., Burge C.B. Classification of introns: U2-type or U12-type. Cell. 1997;91:875–879. doi: 10.1016/S0092-8674(00)80479-1
  32. Stark B.C., Kole R., Bowman E.J., Altman S. Ribonuclease P: an enzyme with an essential RNA component. PNAS. 1978;75:3717–3721. doi: 10.1073/pnas.75.8.3717
  33. Walter P., Blobel G. Signal recognition particle contains a 7S RNA essential for protein translocation across the endoplasmic reticulum. Nature. 1982;299:691–698. doi: 10.1038/299691a0
  34. Lewin R. Surprising discovery with a small RNA. Science. 1982;218:777–778. doi: 10.1126/science.6182614
  35. Bartel D.P. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297. doi: 10.1016/S0092-8674(04)00045-5
  36. Bartel D.P., Chen C.Z. Micromanagers of gene expression: the potentially widespread influence of metazoan microRNAs. Nat. Rev. Genet. 2004;5:396–400. doi: 10.1038/nrg1328
  37. Fragnet L., Kut E., Rasschaert D. Comparative functional study of the viral telomerase RNA based on natural mutations. J. Biol. Chem. 2005;280:23502–23515. doi: 10.1074/jbc.M501163200
  38. Plath K., Mlynarczyk-Evans S., Nusinow D.A., Panning B. Xist RNA and the mechanism of X chromosome inactivation. Annu. Rev. Genet. 2002;36:233–278. doi: 10.1146/annurev.genet.36.042902.092433
  39. Brockdorff N., Ashworth A., Kay G.F., McCabe V.M., Norris D.P., Cooper P.J., Swift S., Rastan S. The product of the mouse Xist gene is a 15 kb inactive X-specific transcript containing no conserved ORF and located in the nucleus. Cell. 1992;71:515–526. doi: 10.1016/0092-8674(92)90519-I
  40. Kelley R.L., Kuroda M.L. Noncoding RNA genes in dosage compensation and imprinting. Cell. 2000;103:9–12. doi: 10.1016/S0092-8674(00)00099-4
  41. Sleutels F., Zwart R., Barlow D.P. The non-coding Air RNA is required for silencing autosomal imprinted genes. Nature. 2002;415:810–813. doi: 10.1038/415810a
  42. Ulitsky I., Bartel D.P. lincRNAs: genomics, evolution, and mechanisms. Cell. 2013;154:26–46. doi: 10.1016/j.cell.2013.06.020
  43. Mutz K.O., Heilkenbrinker A., Lönne M., Walter J.G., Stahl F. Transcriptome analysis using next-generation sequencing. Current Opinion in Biotechnology. 2013;24(1):22–30. doi: 10.1016/j.copbio.2012.09.004
  44. 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. doi: 10.1016/0092-8674(93)90529-Y
  45. Fabian M.R., Sonenberg N., Filipowicz W. Regulation of mRNA translation and stability by microRNAs. Annu. Rev. Biochem. 2010;79:351–379. doi: 10.1146/annurev-biochem-060308-103103
  46. Bissels U., Wild S., Tomiuk S., Holste A., Hafner M., Tuschl T., Bosio A. Absolute quantification of microRNAs by using a universal reference. RNA. 2009;15:2375–2384. doi: 10.1261/rna.1754109
  47. Hamilton A.J., Baulcombe D.C. A species of small antisense RNA in posttranscriptional gene silencing in plants. Science. 1999;286:950–952. doi: 10.1126/science.286.5441.950
  48. 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. doi: 10.1101/gad.203786.112
  49. 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(7441):384–388. doi: 10.1038/nature11993
  50. Memczak S., Jens M., Elefsinioti A., Torti F., Krueger J., Rybak A., Maier L., Mackowiak S.D., Gregersen L.H., Munschauer M. et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495(7441):333–338. doi: 10.1038/nature11928
  51. Valdmanis P.N., Kay M.A. The expanding repertoire of circular RNAs. Mol. Ther. 2013;21(6):1112–1114. doi: 10.1038/mt.2013.101
  52. Lee S.R., Collins K. Starvation-induced cleavage of the tRNA anticodon loop in Tetrahymena thermophila. J. Biol. Chem. 2005;280:42744–42749. doi: 10.1074/jbc.M510356200
  53. Haiser H.J., Karginov F.V., Hannon G.J., Elliot M.A. Developmentally regulated cleavage of tRNAs in the bacterium Streptomyces coelicolor. Nucleic Acids Res. 2008;36:732–741. doi: 10.1093/nar/gkm1096
  54. Chak L.L., Mohammed J., Lai E.C., Tucker-Kellogg G., Okamura K. A deeply conserved, noncanonical miRNA hosted by ribosomal DNA. RNA. 2015;21:375–384. doi: 10.1261/rna.049098.114
  55. Asha S., Soniya E.V. The sRNAome mining revealed existence of unique signature small RNAs derived from 5.8SrRNA from Piper nigrum and other plant lineages. Sci. Rep. 2017;7:41052. doi: 10.1038/srep41052
  56. Chen Z., Sun Y., Yang X., Wu Z., Guo K., Niu X., Wang Q., Ruan J., Bu W., Gao S. Two featured series of rRNA-derived RNA fragments (rRFs) constitute a novel class of small RNAs. PLoS One. 2017;12:e0176458. doi: 10.1371/journal.pone.0176458
  57. Cho J. Transposon-Derived Non-coding RNAs and Their Function in Plants. Front. Plant Sci. 2018;9(600). doi: 10.3389/fpls.2018.00600
  58. Wolf S.F., Schlessinger D. Nuclear metabolism of ribosomal RNA in growing, methionine-limited, and ethionine-treated HeLa cells. Biochemistry (Mosc.) 1977;16:2783–2791. doi: 10.1021/bi00631a031
  59. Natsidis P., Schiffer P.H., Salvador-Martínez I., Telford M.J. Computational discovery of hidden breaks in 28S ribosomal RNAs across eukaryotes and consequences for RNA Integrity Numbers. Sci. Rep. 2019;9(19477). doi: 10.1038/s41598-019-55573-1
  60. Waldron C., Lacroute F. Effect of growth rate on the amounts of ribosomal and transfer ribonucleic acids in yeast. J. Bacteriol. 1975;122:855–865. doi: 10.1128/jb.122.3.855-865.1975
  61. 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. doi: 10.1038/nrm2124
  62. Dieci G., Preti M., Montanini B. Eukaryotic snoRNAs: a paradigm for gene expression flexibility. Genomics. 2009;94:83–88. doi: 10.1016/j.ygeno.2009.05.002
  63. Aravin A., Tuschl T. Identification and characterization of small RNAs involved in RNA silencing. FEBS Lett. 2005;579(26):5830–5840. doi: 10.1016/j.febslet.2005.08.009
  64. Siomi M., 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. doi: 10.1038/nrm3089
  65. Lee Y.S., Shibata Y., Malhotra A., Dutta A. A novel class of small RNAs: tRNA-derived RNA fragments (tRFs). Genes Dev. 2009;23:2639–2649. doi: 10.1101/gad.1837609
  66. Guan L., Grigoriev A. Computational meta-analysis of ribosomal RNA fragments: potential targets and interaction mechanisms. Nucleic Acids Res. 2021;49(7):4085–4103. doi: 10.1093/nar/gkab190
  67. Wilusz J.E., Sunwoo H., Spector D.L. Long noncoding RNAs: functional surprises from the RNA world. Genes Dev. 2009;23(13):1494–1504. doi: 10.1101/gad.1800909
  68. Chen L., Huang C., Wang X., Shan G. Circular RNAs in eukaryotic cells. Curr. Genom. 2015;16(5):312–318. doi: 10.2174/1389202916666150707161554
  69. Gerbi S.A. Expansion segments: regions of variable size that interrupt the universal core secondary structure of ribosomal RNA. In: Ribosomal RNA: Structure, Evolution, Processing and Function in Protein Synthesis. Eds: R.A. Zimmermann and A.E. Dahlberg. Boca Raton, FL: Telford – CRC Press, 1996. P. 71–87.
  70. Armache J.-P., Jarasch A., Anger A.M., Villa E., Becker T., Bhushan S., Jossinet F., Habeck M., Dindar G., Franckenberg S., et al. Cryo-EM structure and rRNA model of a translating eukaryotic 80S ribosome at 5.5-A resolution. PNAS. 2010;107:19748–19753. doi: 10.1073/pnas.1009999107
  71. Anger A.M., Armache J.-P., Berninghausen O., Habeck M., Subklewe M., Wilson D.N., Beckmann R. Structures of the human and Drosophila 80S ribosome. Nature. 2013;497:80–85. doi: 10.1038/nature12104
  72. Fujii K., Susanto T.T., Saurabh S., Barna M. Decoding the function of expansion segments in ribosomes. Mol. Cell. 2018;72:1013–1020. doi: 10.1016/j.molcel.2018.11.023
  73. Chan P.P., Lowe T.M. GtRNAdb: a database of transfer RNA genes detected in genomic sequence. Nucleic Acids Res. 2009;37. P. D93–D97. doi: 10.1093/nar/gkn787
  74. Parisien M., Wang X., Pan T. Diversity of human tRNA genes from the 1000-genomes project. RNA Biol. 2013;10:1853–1867. doi: 10.4161/rna.27361
  75. Orioli A. tRNA biology in the omics era: stress signalling dynamics and cancer progression. Bioessays. 2017;39:1600158. doi: 10.1002/bies.201600158
  76. Kirchner S., Ignatova Z. Emerging roles of tRNA in adaptive translation, signalling dynamics and disease. Nat. Rev. Genet. 2015;16:98–112. doi: 10.1038/nrg3861
  77. Huang S.Q., Sun B., Xiong Z.P., Shu Y., Zhou H.H., Zhang W., Xiong J., Li Q. The dysregulation of tRNAs and tRNA derivatives in cancer. J. Exp. Clin. Cancer Res. 2018;37:101. doi: 10.1186/s13046-018-0745-z
  78. Santos M., Fidalgo A., Varanda A.S., Oliveira C., Santos M.A.S. tRNA deregulation and its consequences in cancer. Trends Mol. Med. 2019;25:853–865. doi: 10.1016/j.molmed.2019.05.011
  79. Gomez-Roman N., Grandori C., Eisenman R.N., White R.J. Direct activation of RNA polymerase III transcription by c-Myc. Nature. 2003;421:290–294. doi: 10.1038/nature01327
  80. Felton-Edkins Z.A., Fairley J.A., Graham E.L., Johnston I.M., White R.J., Scott P.H. The mitogen-activated protein (MAP) kinase ERK induces tRNA synthesis by phosphorylating TFIIIB. EMBO J. 2003;22:2422–2432. doi: 10.1093/emboj/cdg240
  81. Wei Y., Tsang C.K., Zheng X.F. Mechanisms of regulation of RNA polymerase III-dependent transcription by TORC1. EMBO J. 2009;28:2220–2230. doi: 10.1038/emboj.2009.179
  82. Truitt M.L., Ruggero D. New frontiers in translational control of the cancer genome. Nat. Rev. Cancer. 2016;16:288–304. doi: 10.1038/nrc.2016.27
  83. Beznosková P., Bidou L., Namy O., Valášek L.S. Increased expression of tryptophan and tyrosine tRNAs elevates stop codon readthrough of reporter systems in human cell lines. Nucleic Acids Res. 2021;49(9):5202–5215. doi: 10.1093/nar/gkab315
  84. Goodenbour J.M., Pan T. Diversity of tRNA genes in eukaryotes. Nucleic Acids Res. 2006;34:6137–6146. doi: 10.1093/nar/gkl725
  85. Mahlab S., Tuller T., Linial M. Conservation of the relative tRNA composition in healthy and cancerous tissues. RNA. 2012;18:640–652. doi: 10.1261/rna.030775.111
  86. Pavon-Eternod M., Gomes S., Geslain R., Dai Q., Rosner M.R., Pan T. tRNA over-expression in breast cancer and functional consequences. Nucleic Acids Res. 2009;37:7268–7280. doi: 10.1093/nar/gkp787
  87. Gingold H., Tehler D., Christoffersen N.R., Nielsen M.M., Asmar F., Kooistra S.M., Christophersen N.S., Christensen L.L., Borre M., Sorensen K.D. et al. A dual program for translation regulation in cellular proliferation and differentiation. Cell. 2014;158:1281–1292. doi: 10.1016/j.cell.2014.08.011
  88. Goodarzi H., Nguyen H.C.B., Zhang S., Dill B.D., Molina H., Tavazoie S.F. Modulated expression of specific tRNAs drives gene expression and cancer progression. Cell. 2016;165:1416–1427. doi: 10.1016/j.cell.2016.05.046
  89. Jackson R.J., Hellen C.U., Pestova T.V. Termination and post-termination events in eukaryotic translation. Adv. Protein Chem. Struct. Biol. 2012;86:45–93. doi: 10.1016/B978-0-12-386497-0.00002-5
  90. Valasek L.S., Zeman J., Wagner S., Beznoskova P., Pavlikova Z., Mohammad M.P., Hronova V., Herrmannova A., Hashem Y., Gunisova S. Embraced by eIF3: structural and functional insights into the roles of eIF3 across the translation cycle. Nucleic Acids Res. 2017;45:10948–10968. doi: 10.1093/nar/gkx805
  91. Dabrowski M., Bukowy-Bieryllo Z., Zietkiewicz E. Translational readthrough potential of natural termination codons in eucaryotes–The impact of RNA sequence. RNA Biol. 2015;12:950–958. doi: 10.1080/15476286.2015.1068497
  92. Schueren F., Thoms S. Functional translational readthrough: a systems biology perspective. PLoS Genet. 2016;12:e1006196. doi: 10.1371/journal.pgen.1006196
  93. Schueren F., Lingner T., George R., Hofhuis J., Dickel C., Gartner J., Thoms S. Peroxisomal lactate dehydrogenase is generated by translational readthrough in mammals. Elife. 2014;3:e03640. doi: 10.7554/eLife.03640
  94. Loughran G., Chou M.Y., Ivanov I.P., Jungreis I., Kellis M., Kiran A.M., Baranov P.V., Atkins J.F. Evidence of efficient stop codon readthrough in four mammalian genes. Nucleic Acids Res. 2014;42:8928–8938. doi: 10.1093/nar/gku608
  95. Avcilar-Kucukgoze I., Kashina A. Hijacking tRNAs from Translation: Regulatory Functions of tRNAs in Mammalian Cell Physiology. Front. Mol. Biosci. 2020;7(610617). doi: 10.3389/fmolb.2020.610617
  96. Torres A.G. Enjoy the Silence: Nearly Half of Human tRNA Genes Are Silent. Bioinformatics Biol. Insights. 2019;13. doi: 10.1177/1177932219868454
  97. Soma A., Onodera A., Sugahara J., Kanai A., Yachie N., Tomita M., Kawamura F., Sekine Y. Permuted tRNA Genes Expressed via a Circular RNA Intermediate in Cyanidioschyzon Merolae. Science. 2007;318(5849):450–453. doi: 10.1126/science.1145718
  98. Randau L., Münch R., Hohn M.J., Jahn D., Söll D. Nanoarchaeum Equitans Creates Functional tRNAs from Separate Genes for Their 5′- and 3′-halves. Nature. 2005;433:537–541. doi: 10.1038/nature03233
  99. Fujishima K., Sugahara J., Kikuta K., Hirano R., Sato A., Tomita M., Kanai A. Tri-split tRNA Is a Transfer RNA Made from 3 Transcripts that Provides Insight into the Evolution of Fragmented tRNAs in Archaea. PNAS. 2009;106(8):2683–2687. doi: 10.1073/pnas.0808246106
  100. Cheng J., Kapranov P., Drenkow J., Dike S., Brubaker S., Patel S., Long J., Stern D., Tammana H., Helt G. et al. Transcriptional maps of 10 human chromosomes at 5-nucleotide resolution. Science. 2005;308:1149–1154. doi: 10.1126/science.1108625
  101. Mattick J.S., Makunin I.V. Non-coding RNA. Hum. Mol. Genet. 2006;15:R17–R29. doi: 10.1093/hmg/ddl046
  102. Huttenhofer A., Schattner P., Polacek N. Noncoding RNAs: hope or hype? Trends Genet. 2005;21:289–297. doi: 10.1016/j.tig.2005.03.007
  103. Storz G., Altuvia S., Wassarman K.M. An abundance of RNA regulators. Annu. Rev. Biochem. 2005;74:199–217. doi: 10.1146/annurev.biochem.74.082803.133136
  104. Matera A.G., Wang Z. A day in the life of the spliceosome. Nat. Rev. Mol. Cell Biol. 2014;15(2):108–121. doi: 10.1038/nrm3742
  105. Szymanski M., Barciszewska M.Z., Zywicki M., Barciszewski J. Noncoding RNA transcripts. J. Appl. Genet. 2003;44:1–19.
  106. 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(3):209–220. doi: 10.1038/nrm2124
  107. Bohnsack M.T., Sloan K.E. Modifications in small nuclear RNAs and their roles in spliceosome assembly and function. Biological Chemistr. 2018;399(11):1265–1276. doi: 10.1515/hsz-2018-0205
  108. Will C.L., Lührmann R. Spliceosome structure and function. Cold Spring Harb. Perspect. Biol. 2011;3. doi: 10.1101/cshperspect.a003707
  109. Hoskins A.A., Moore M.J. The spliceosome: a flexible, reversible macromolecular machine. Trends Biochem. Sci. 2012;37:179–188. doi: 10.1016/j.tibs.2012.02.009
  110. Karijolich J., Yu Y.T. Spliceosomal snRNA modifications and their function. RNA Biol. 2010;7:192–204. doi: 10.4161/rna.7.2.11207
  111. Morais P., Adachi H., Yu Y.T. Spliceosomal snRNA Epitranscriptomics. Front Genet. 2021;12(652129). doi: 10.3389/fgene.2021.652129
  112. Stein C.A., Castanotto D. FDA-approved oligonucleotide therapies in 2017. Mol. Ther. 2017;25:1069–1075. doi: 10.1016/j.ymthe.2017.03.023
  113. Rüger J., Ioannou S., Castanotto D., Stein C.A. Oligonucleotides to the (gene) rescue: FDA approvals 2017-2019. Trends Pharmacol. Sci. 2020;41:27–41. doi: 10.1016/j.tips.2019.10.009
  114. Eliceiri G.L. Small nucleolar RNAs. Cell. Mol. Life Sci. 1999;56:22–31. doi: 10.1007/s000180050003
  115. 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. doi: 10.1038/nrm2124
  116. Boivin V., Faucher-Giguère L., Scott M., Abou-Elela S. The cellular landscape of mid-size noncoding RNA. Wiley Interdiscip Rev RNA. 2019;10:e1530. doi: 10.1002/wrna.1530
  117. Bergeron D., Fafard-Couture É., Scott M.S. Small nucleolar RNAs: continuing identification of novel members and increasing diversity of their molecular mechanisms of action. Biochem. Soc. Trans. 2020;48:645–656. doi: 10.1042/BST20191046
  118. Kiss T. Small nucleolar RNA-guided post-transcriptional modification of cellular RNAs. EMBO J. 2001;20:3617–3622. doi: 10.1093/emboj/20.14.3617
  119. Bergeron D., Laforest C., Carpentier S., Calvé A., Fafard-Couture É., Deschamps-Francoeur G., Scott M.S. SnoRNA copy regulation affects family size, genomic location and family abundance levels. BMC Genomics. 2021;22(414). doi: 10.1186/s12864-021-07757-1
  120. Deschamps-Francoeur G., Garneau D., Dupuis-Sandoval F., Roy A., Frappier M., Catala M., Couture S., Barbe-Marcoux M., Abou-Elela S., Scott M.S. Identification of discrete classes of small nucleolar RNA featuring different ends and RNA binding protein dependency. Nucleic Acids Res. 2014;42:10073–10085. doi: 10.1093/nar/gku664
  121. Marmier-Gourrier N., Cléry A., Senty-Ségault V., Charpentier B., Schlotter F., Leclerc F., Fournier R., Branlant C. A structural, phylogenetic, and functional study of 15.5-kD/Snu13 protein binding on U3 small nucleolar RNA. RNA. 2003;9:821–838. doi: 10.1261/rna.2130503
  122. Lestrade L., Weber M.J. snoRNA-LBME-db, a comprehensive database of human H/ACA and C/D box snoRNAs. Nucleic Acids Res. 2006;34:D158. doi: 10.1093/nar/gkj002
  123. Ganot P., Caizergues-Ferrer M., Kiss T. The family of box ACA small nucleolar RNAs is defined by an evolutionarily conserved secondary structure and ubiquitous sequence elements essential for RNA accumulation. Genes Dev. 1997;11:941–956. doi: 10.1101/gad.11.7.941
  124. Ganot P., Bortolin M.L., Kiss T. Site-specific pseudouridine formation in preribosomal RNA is guided by small nucleolar RNAs. Cell. 1997;89:799–809. doi: 10.1016/S0092-8674(00)80263-9
  125. Brown J.W., Marshall D.F., Echeverria M. Intronic noncoding RNAs and splicing. Trends Plant Sci. 2008;13:335–342. doi: 10.1016/j.tplants.2008.04.010
  126. Chanfreau G., Legrain P., Jacquier A. Yeast RNase III as a key processing enzyme in small nucleolar RNAs metabolism. J. Mol. Biol. 1998;284:975–988. doi: 10.1006/jmbi.1998.2237
  127. Petfalski E., Dandekar T., Henry Y., Tollervey D. Processing of the precursors to small nucleolar RNAs and rRNAs requires common components. Mol. Cell. Biol. 1998;18:1181–1189. doi: 10.1128/MCB.18.3.1181
  128. Qu L.H., Henras A., Lu Y.J., Zhou H., Zhou W.X., Zhu Y.Q., Zhao J., Henry Y., Caizergues-Ferrer M., Bachellerie J.P. Seven novel methylation guide small nucleolar RNAs are processed from a common polycistronic transcript by Rat1p and RNase III in yeast. Mol. Cell. Biol. 1999;19:1144–1158. doi: 10.1128/MCB.19.2.1144
  129. Dupuis-Sandoval F., Poirier M., Scott M.S. The emerging landscape of small nucleolar RNAs in cell biology. Wiley Interdiscip. Rev. RNA. 2015;6:381–397. doi: 10.1002/wrna.1284
  130. Bratkovič T., Bozič J., Rogelj B. Functional diversity of small nucleolar RNAs. Nucleic Acids Res. 2020;48:1627–1651. doi: 10.1093/nar/gkz1140
  131. Falaleeva M., Welden J.R., Duncan M.J., Stamm S. C/D-box snoRNAs form methylating and non-methylating ribonucleoprotein complexes: old dogs show new tricks. BioEssays. 2017;39. doi: 10.1002/bies.201600264
  132. Shabalina S.A., Koonin E.V. Origins and evolution of eukaryotic RNA interference. Trends Ecol. Evol. (Amst.). 2008;23(10):578–587. doi: 10.1016/j.tree.2008.06.005
  133. Wilson R.C., Doudna J.A. Molecular mechanisms of RNA interference. Annu. Rev. Biophys. 2013;42:217–239. doi: 10.1146/annurev-biophys-083012-130404
  134. Fabian M.R., Sonenberg N. The mechanics of miRNA-mediated gene silencing: a look under the hood of miRISC. Nature Structural & Molecular Biology. 2012;19(6):586–593. doi: 10.1038/nsmb.2296
  135. Carthew R.W., Sontheimer E.J. Origins and Mechanisms of miRNAs and siRNAs. Cell. 2009;136(4):642–655. doi: 10.1016/j.cell.2009.01.035
  136. Lu M., Zhang Q., Deng M., Miao J., Guo Y., Gao W., Cui Q. An analysis of human microRNA and disease associations. PLoS ONE. 2008;3(10):e3420. doi: 10.1371/journal.pone.0003420
  137. Kubowicz P., Zelaszczyk D., Pekala E. RNAi in clinical studies. Curr. Med. Chem. 2013;20:1801–1816. doi: 10.2174/09298673113209990118
  138. Martinez T., Wright N., López-Fraga M., Jiménez A.I., Pañeda C. Silencing human genetic diseases with oligonucleotide-based therapies. Hum. Genet. 2013;132:481–493. doi: 10.1007/s00439-013-1288-1
  139. Ramachandran P.V., Ignacimuthu S. RNA interference–a silent but an efficient therapeutic tool. Appl. Biochem. Biotechnol. 2013;169:1774–1789. doi: 10.1007/s12010-013-0098-1
  140. Davidson B.L., McCray P.B. Current prospects for RNA interference-based therapies. Nat. Rev. Genet. 2011;12:329–340. doi: 10.1038/nrg2968
  141. Setten R.L., Rossi J.J., Han S. The current state and future directions of RNAi-based therapeutics. Nature Reviews Drug Discovery. 2019;18(6):421–446. doi: 10.1038/s41573-019-0017-4
  142. Carthew R.W., Sontheimer E.J. Origins and Mechanisms of miRNAs and siRNAs. Cell. 2009;136(4):642–655. doi: 10.1016/j.cell.2009.01.035
  143. Saini H.K., Griffiths-Jones S., Enright A.J. Genomic analysis of human microRNA transcripts. PNAS. 2007;104(45):17719–17724. doi: 10.1073/pnas.0703890104
  144. Han J., Lee Y., Yeom K.-H., Nam J.-W., Heo I., Rhee J.-K., Sohn S.Y., Cho Y., Zhang B.-T., Kim V.N. Molecular Basis for the Recognition of Primary microRNAs by the Drosha-DGCR8 Complex. Cell. 2006;125(5):887–901. doi: 10.1016/j.cell.2006.03.043
  145. Lund E., Dahlberg J.E. Substrate selectivity of exportin 5 and Dicer in the biogenesis of microRNAs. Cold Spring Harb. Symp. Quant. Biol. 2006;71:59–66. doi: 10.1101/sqb.2006.71.050
  146. Schwarz D.S., Hutvágner G., Du T., Xu Z., Aronin N., Zamore P.D. Asymmetry in the assembly of the RNAi enzyme complex. Cell. 2003;115(2):199–208. doi: 10.1016/S0092-8674(03)00759-1
  147. MacRae I.J., Ma E., Zhou M., Robinson C.V., Doudna J.A. In vitro reconstitution of the human RISC-loading complex. PNAS. 2008;105(2):512–517. doi: 10.1073/pnas.0710869105
  148. Eulalio A., Huntzinger E., Izaurralde E. GW182 interaction with Argonaute is essential for miRNA-mediated translational repression and mRNA decay. Nature Structural & Molecular Biology. 2008;15(4):346–353. doi: 10.1038/nsmb.1405
  149. Flynt A.S., Lai E.C. Biological principles of microRNA-mediated regulation: shared themes amid diversity. Nat. Rev. Genet. 2008;9:831–842. doi: 10.1038/nrg2455
  150. Axtell M.J., Westholm J.O., Lai E.C. Vive la différence: biogenesis and evolution of microRNAs in plants and animals. Genome Biol. 2011;12:221. doi: 10.1186/gb-2011-12-4-221
  151. Westholm J.O., Lai E.C. Mirtrons: microRNA biogenesis via splicing. Biochimie. 2011;93:1897–1904. doi: 10.1016/j.biochi.2011.06.017
  152. Czech B., Hannon G.J. Small RNA sorting: matchmaking for Argonautes. Nat. Rev. Genet. 2010;12:19–31. doi: 10.1038/nrg2916
  153. 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. doi: 10.1261/rna.2183803
  154. Yang J.S., Lai E.C. Alternative miRNA biogenesis pathways and the interpretation of core miRNA pathway mutants. Mol. Cell. 2011;43:892–903. doi: 10.1016/j.molcel.2011.07.024
  155. Okamura K., Hagen J.W., Duan H., Tyler D.M., Lai E.C. The mirtron pathway generates microRNA-class regulatory RNAs in Drosophila. Cell. 2007;130:89–100. doi: 10.1016/j.cell.2007.06.028
  156. Ruby J.G., Jan C.H., Bartel D.P. Intronic microRNA precursors that bypass Drosha processing. Nature. 2007;448:83–86. doi: 10.1038/nature05983
  157. Flynt A.S., Chung W.J., Greimann J.C., Lima C.D., Lai E.C. microRNA biogenesis via splicing and exosome-mediated trimming in Drosophila. Mol. Cell. 2010;38:900–907. doi: 10.1016/j.molcel.2010.06.014
  158. Ladewig E., Okamura K., Flynt A.S., Westholm J.O., Lai E.C. Discovery of hundreds of mirtrons in mouse and human small RNA data. Genome Res. 2012;22(9):1634–1645. doi: 10.1101/gr.133553.111
  159. Wen J., Ladewig E., Shenker S., Mohammed J., Lai E.C. Analysis of Nearly One Thousand Mammalian Mirtrons Reveals Novel Features of Dicer Substrates. PLoS Comput. Biol. 2015;11(9):e1004441. doi: 10.1371/journal.pcbi.1004441
  160. Westholm J.O., Ladewig E., Okamura K., Robine N., Lai E.C. Common and distinct patterns of terminal modifications to mirtrons and canonical microRNAs. RNA. 2012;18:177–192. doi: 10.1261/rna.030627.111
  161. Lai E.C. microRNAs: Runts of the genome assert themselves. Curr. Biol. 2003;13:R925–R936. doi: 10.1016/j.cub.2003.11.017
  162. Doench J.G., Sharp P.A. Specificity of microRNA target selection in translational repression. Genes Dev. 2004;18(5):504–511. doi: 10.1101/gad.1184404
  163. Brennecke J., Stark A., Russell R.B., Cohen S.M. Principles of microRNA-target recognition. PLoS Biol. 2005;3(3):e85. doi: 10.1371/journal.pbio.0030085
  164. Lewis B.P., Burge C.B., Bartel D.P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120(1):15–20. doi: 10.1016/j.cell.2004.12.035
  165. Bartel D.P. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–233. doi: 10.1016/j.cell.2009.01.002
  166. Krek A., Grün D., Poy M.N., Wolf R., Rosenberg L., Epstein E.J., MacMenamin P., da Piedade I., Gunsalus K.C., Stoffel M., Rajewsky N. Combinatorial microRNA target predictions. Nat. Genet. 2005;37(5):495–500. doi: 10.1038/ng1536
  167. Friedman R.C., Farh K.K., Burge C.B., Bartel D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19(1):92–105. doi: 10.1101/gr.082701.108
  168. Simkin A., Geissler R., McIntyre A.B.R., Grimson A. Evolutionary dynamics of microRNA target sites across vertebrate evolution. PLoS Genet. 2020;16(2):e1008285. doi: 10.1371/journal.pgen.1008285
  169. Zamore P.D., Tuschl T., Sharp P.A., Bartel D.P. RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell. 2000;101:25–33. doi: 10.1016/S0092-8674(00)80620-0
  170. Elbashir S.M., Lendeckel W., Tuschl T. RNA interference is mediated 1- and 22-nucleotide RNAs. Genes Dev. 2001;15:188–200. doi: 10.1101/gad.862301
  171. Sijen T., Plasterk R.H. Transposon silencing in the Caenorhabditis elegans germline by natural RNAi. Nature. 2003;426:310–314. doi: 10.1038/nature02107
  172. Shi H., Djikeng A., Tschudi C., Ullu E. Argonaute protein in the early divergent eukaryote Trypanosoma brucei: Control of small interfering RNA accumulation and retroposon transcript abundance. Mol. Cell. Biol. 2004;24:420–427. doi: 10.1128/MCB.24.1.420-427.2004
  173. Lippman Z., Martienssen R. The role of RNA interference in heterochromatic silencing. Nature. 2004;431:364–370. doi: 10.1038/nature02875
  174. Peragine A., Yoshikawa M., Wu G., Albrecht H.L., Poethig R.S. SGS3 and SGS2/SDE1/RDR6 are required for juvenile development and the production of trans-acting siRNAs in Arabidopsis. Genes & Dev. 2004;18:2368–2379. doi: 10.1101/gad.1231804
  175. Borsani O., Zhu J., Verslues P.E., Sunkar R., Zhu J.K. Endogenous siRNAs derived from a pair of natural cis-antisense transcripts regulate salt tolerance in Arabidopsis. Cell. 2005;123:1279–1291. doi: 10.1016/j.cell.2005.11.035
  176. Ambros V., Lee R.C., Lavanway A., Williams P.T., Jewell D. MicroRNAs and other tiny endogenous RNAs in C. elegans. Curr. Biol. 2003;13:807–818. doi: 10.1016/S0960-9822(03)00287-2
  177. Chen P.Y., Manninga H., Slanchev K., Chien M., Russo J.J., Ju J., Sheridan R., John B., Marks D.S., Gaidatzis D., et al. The developmental miRNA profiles of zebrafish as determined by small RNA cloning. Genes & Dev. 2005;19:1288–1293. doi: 10.1101/gad.1310605
  178. Aravin A.A. The small RNA profile during Drosophila melanogaster development. Dev. Cell. 2003;5:337–350. doi: 10.1016/S1534-5807(03)00228-4
  179. Moldovan D., Spriggs A., Dennis E.S., Wilson I.W. The hunt for hypoxia responsive natural antisense short interfering RNAs. Plant Signaling & Behavior. 2010;5:247–251. doi: 10.4161/psb.5.3.10548
  180. Katiyar-Agarwal S., Morgan R., Dahlbeck D., Borsani O., Villegas A., Zhu J.-K., Staskawicz B.J., Jin H. A pathogen-inducible endogenous siRNA in plant immunity. PNAS. 2006;103(47):18002–18007. doi: 10.1073/pnas.0608258103
  181. Held M.A., Penning B., Brandt A.S., Kessans S.A., Yong W., Scofield S.R., Carpita N.C. Small-interfering RNAs from natural antisense transcripts derived from a cellulose synthase gene modulate cell wall biosynthesis in barley. PNAS. 2008;105(51):20534–20539. doi: 10.1073/pnas.0809408105
  182. Zhang X., Xia J., Lii Y.E., Barrera-Figueroa B.E., Zhou X., Gao S., Lu L., Niu D., Chen Z., Leung C. et al.Genome-wide analysis of plant nat-siRNAs reveals insights into their distribution, biogenesis and function. Genome Biology. 2012;13(20):R20. doi: 10.1186/gb-2012-13-3-r20
  183. Watanabe T., Takeda A., Tsukiyama T., Mise K., Okuno T., Sasaki H., Minami N., Imai H. Identification and characterization of two novel classes of small RNAs in the mouse germline: retrotransposon-derived siRNAs in oocytes and germline small RNAs in testes. Genes Dev. 2006;20(13):1732–1743. doi: 10.1101/gad.1425706
  184. Aravin A., Gaidatzis D., Pfeffer S., Lagos-Quintana M., Landgraf P., Iovino N., Morris P., Brownstein M.J., Kuramochi-Miyagawa S., Nakano T.et al. A novel class of small RNAs bind to MILI protein in mouse testes. Nature. 2006;442:203–207. doi: 10.1038/nature04916
  185. Girard A., Sachidanandam R., Hannon G.J., Carmell M.A. A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature. 2006;442(7099):199–202. doi: 10.1038/nature04917
  186. Lau N.C., Seto A.G., Kim J., Kuramochi-Miyagawa S., Nakano T., Bartel D.P., Kingston R.E. Characterization of the piRNA Complex from Rat Testes. Science. 2006;313:363–367. doi: 10.1126/science.1130164
  187. Grivna S.T., Beyret E., Wang Z., Lin H. A novel class of small RNAs in mouse spermatogenic cells. Genes Dev. 2006;20:1709–1714. doi: 10.1101/gad.1434406
  188. Malone C.D., Hannon G.J. Small RNAs as guardians of the genome. Cell. 2009;136(4):656–668. doi: 10.1016/j.cell.2009.01.045
  189. 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(21):2361–2373. doi: 10.1101/gad.203786.112
  190. Gou L.T., Dai P., Yang J.H., Xue Y., Hu Y.P., Zhou Y., Kang J.Y., Wang X., Li H., Hua M.M. et al. Pachytene piRNAs instruct massive mRNA elimination during late spermiogenesis. Cell Res. 2014;24:680–700. doi: 10.1038/cr.2014.41
  191. Zhang P., Kang J.Y., Gou L.T., Wang J., Xue Y., Skogerboe G., Dai P., Huang D.W., Chen R., Fu X.D. et al. MIWI and piRNAmediated cleavage of messenger RNAs in mouse testes. Cell Res. 2015;25:193–207. doi: 10.1038/cr.2015.4
  192. Goh W.S.S., Falciatori I., Tam O.H., Burgess R., Meikar O., Kotaja N., Hammell M., Hannon G.J. piRNA-directed cleavage of meiotic transcripts regulates spermatogenesis. Genes Dev. 2015;29:1032–1044. doi: 10.1101/gad.260455.115
  193. Watanabe T., Cheng E., Zhong M., Lin H. Retrotransposons and pseudogenes regulate mRNAs and lncRNAs via the piRNA pathway in the germline. Genome Res. 2015;25:368–380. doi: 10.1101/gr.180802.114
  194. Vourekas A., Alexiou P., Vrettos N., Maragkakis M., Mourelatos Z. Sequence-dependent but not sequence-specific piRNA adhesion traps mRNAs to the germ plasm. Nature. 2016;531:390–394. doi: 10.1038/nature17150
  195. Shen E.Z., Chen H., Ozturk A.R., Tu S., Shirayama M., Tang W., Ding Y.H., Dai S.Y., Weng Z., Mello C.C. Identification of piRNA binding sites reveals the Argonaute regulatory landscape of the C. elegans germline. Cell. 2018;172:937–951.e18. doi: 10.1016/j.cell.2018.02.002
  196. Zhang D., Tu S., Stubna M., Wu W.S., Huang W.C., Weng Z., Lee H.C. The piRNA targeting rules and the resistance to piRNA silencing in endogenous genes. Science. 2018;359:587–592. doi: 10.1126/science.aao2840
  197. Dai P., Wang X., Gou L.T., Li Z.T., Wen Z., Chen Z.G., Hua M.M., Zhong A., Wang L., Su H., Wan H., Qian K., Liao L., Li J., Tian B., Li D., Fu X.D., Shi H.J., Zhou Y., Liu M.F. A Translation-Activating Function of MIWI/piRNA during Mouse Spermiogenesis. Cell. 2019;179(7):1566–1581.E16. doi: 10.1016/j.cell.2019.11.022
  198. Dai P., Wang X., Liu MF. A dual role of the PIWI/piRNA machinery in regulating mRNAs during mouse spermiogenesis. Sci. China Life Sci. 2020;63:447–449. doi: 10.1007/s11427-020-1632-5
  199. Brennecke J., Aravin A.A., Stark A., Dus M., Kellis M., Sachidanandam R., Hannon G.J. Discrete small RNA-generating loci as master regulators of transposon activity in Drosophila. Cell. 2007;128:1089–1103. doi: 10.1016/j.cell.2007.01.043
  200. Ozata D.M., Gainetdinov I., Zoch A., O’Carroll D., Zamore P.D. PIWI-interacting RNAs: small RNAs with big functions. Nat. Rev. Genet. 2019;20:89–108. doi: 10.1038/s41576-018-0073-3
  201. Seto A.G., Kingston R.E., Lau N.C. The Coming of Age for Piwi Proteins. Molecular Cell. 2007;26(5):603–609. doi: 10.1016/j.molcel.2007.05.021
  202. Jochl C., Rederstorff M., Hertel J., Stadler P.F., Hofacker I.L., Schrettl M., Haas H., Huttenhofer A. Small ncRNA transcriptome analysis from Aspergillus fumigatus suggests a novel mechanism for regulation of protein synthesis. Nucleic Acids Res. 2008;36:2677–2689. doi: 10.1093/nar/gkn123
  203. Li Y., Zhang Y., Liu M. Knockout Gene-Based Evidence for PIWI-Interacting RNA Pathway in Mammals. Front. Cell Dev. Biol. 2021;9:681188. doi: 10.3389/fcell.2021.681188
  204. Su J.F., Concilla A., Zhang D.Z., Zhao F., Shen F.F., Zhang H., Zhou F.Y. PIWI-interacting RNAs: Mitochondria-based biogenesis and functions in cancer. Genes Dis. 2020;8(5):603–622. doi: 10.1016/j.gendis.2020.09.006
  205. 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. doi: 10.1261/rna.2000810
  206. Ivanov P., Emara M.M., Villen J., Gygi S.P., Anderson P. Angiogenin-induced tRNA fragments inhibit translation initiation. Mol. Cell. 2011;43:613–623. doi: 10.1016/j.molcel.2011.06.022
  207. Gebetsberger J., Zywicki M., Kunzi A., Polacek N. tRNA-Derived Fragments Target the Ribosome and Function as Regulatory Non-coding RNA in Haloferax Volcanii. Archaea. 2012;2012. Article ID 260909. doi: 10.1155/2012/260909
  208. Speer J., Gehrke C.W., Kuo K.C., Waalkes T.P., Borek E. tRNA Breakdown Products as Markers for Cancer. Cancer. 1979;44(6):2120–2123. doi: 10.1002/1097-0142(197912)44:6<2120::AID-CNCR2820440623>3.0.CO;2-6
  209. Balatti V., Nigita G., Veneziano D., Drusco A., Stein G.S., Messier T.L., Farina N.H., Lian J.B., Tomasello L., Liu C.G. et al. tsRNA signatures in cancer. PNAS. 2017;114:8071–8076. doi: 10.1073/pnas.1706908114
  210. Guzman N., Agarwal K., Asthagiri D., Yu L., Saji M., Ringel M.D., Paulaitis M.E. Breast cancer-specific miR signature unique to extracellular vesicles includes “microRNA-like” tRNA fragments. Mol. Cancer Res. 2015:13:891–901. doi: 10.1158/1541-7786.MCR-14-0533
  211. Keam S.P., Hutvagner G. tRNA-Derived Fragments (tRFs): Emerging New Roles for an Ancient RNA in the Regulation of Gene Expression. Life (Basel). 2015;5(4):1638–1651. doi: 10.3390/life5041638
  212. Goodarzi H., Liu X., Nguyen H.C., Zhang S., Fish L., Tavazoie S.F. Endogenous tRNA-derived fragments suppress breast cancer progression via YBX1 displacement. Cell. 2015;161:790–802. doi: 10.1016/j.cell.2015.02.053
  213. Anderson P., Ivanov P. tRNA fragments in human health and disease. FEBS Lett. 2014;588:4297–4304. doi: 10.1016/j.febslet.2014.09.001
  214. Kumar P., Anaya J., Mudunuri S.B., Dutta A. Meta-analysis of tRNA derived RNA fragments reveals that they are evolutionarily conserved and associate with AGO proteins to recognize specific RNA targets. BMC Biol. 2014;12:78. doi: 10.1186/s12915-014-0078-0
  215. Karaiskos S., Grigoriev A. Dynamics of tRNA fragments and their targets in aging mammalian brain. F1000Res. 2016;5:2758. doi: 10.12688/f1000research.10116.1
  216. Jin F., Guo Z. Emerging role of a novel small non-coding regulatory RNA: tRNA-derived small RNA. ExRNA. 2019;1(39). doi: 10.1186/s41544-019-0036-7
  217. Guzzi N., Bellodi C. Novel insights into the emerging roles of tRNA-derived fragments in mammalian development. RNA Biol. 2020;17:1214–1222. doi: 10.1080/15476286.2020.1732694
  218. Megel C., Hummel G., Lalande S., Ubrig E., Cognat V., Morelle G., Salinas-Giegé T., Duchêne A.-M., Maréchal-Drouard L. Plant RNases T2, but not Dicer-like proteins, are major players of tRNA-derived fragments biogenesis. Nucleic Acids Res. 2019;47:941–952. doi: 10.1093/nar/gky1156
  219. Zheng L.L., Xu W.L., Liu S., Sun W.J., Li J.H., Wu J., Yang J.-H., Qu L.-H. tRF2Cancer: a web server to detect tRNA-derived small RNA fragments (tRFs) and their expression in multiple cancers. Nucleic Acids Res. 2016;44:W185–W193. doi: 10.1093/nar/gkw414
  220. Yu M., Lu B., Zhang J., Ding J., Liu P., Lu Y. tRNA-derived RNA fragments in cancer: current status and future perspectives. J. Hematol Oncol. 2020;13:121. doi: 10.1186/s13045-020-00955-6
  221. Thompson D.M., Lu C., Green P.J., Parker R. tRNA cleavage is a conserved response to oxidative stress in eukaryotes. RNA. 2008;14(10):2095–2103. doi: 10.1261/rna.1232808
  222. Shen Y., Yu X., Zhu L., Li T., Yan Z., Guo J. Transfer RNA-derived fragments and tRNA halves: biogenesis, biological functions and their roles in diseases. J. Cell Mol. Med. 2018;96:1167–76. doi: 10.1007/s00109-018-1693-y
  223. Cho H, Lee W, Kim GW, Lee SH, Moon JS, Kim M, Kim HS, Oh JW. Regulation of La/SSB-dependent viral gene expression by pre-tRNA 3' trailer-derived tRNA fragments. Nucleic Acids Res. 2019;47:9888–9901. doi: 10.1093/nar/gkz732
  224. Ivanov P., Emara M.M, Villen J., Gygi S.P, Anderson P. Angiogenin-induced tRNA fragments inhibit translation initiation. Mol Cell. 2011;43:613–623. doi: 10.1016/j.molcel.2011.06.022
  225. Guan L., Karaiskos S., Grigoriev A. Inferring targeting modes of Argonaute-loaded tRNA fragments. RNA Biol. 2020;17:1070–1080. doi: 10.1080/15476286.2019.1676633
  226. Kuscu C., Kumar P., Kiran M., Su Z., Malik A., Dutta A. tRNA fragments (tRFs) guide Ago to regulate gene expression post-transcriptionally in a Dicer-independent manner. RNA. 2018;24:1093–1105. doi: 10.1261/rna.066126.118
  227. Hasler D., Lehmann G., Murakawa Y., Klironomos F., Jakob L., Grasser F.A., Rajewsky N., Landthaler M., Meister G. The lupus autoantigen La prevents mis-channeling of tRNA fragments into the human microRNA pathway. Mol. Cell. 2016;63:110–124. doi: 10.1016/j.molcel.2016.05.026
  228. Anger A.M., Armache J.P., Berninghausen O., Habeck M., Subklewe M., Wilson D.N., Beckmann R. Structures of the human and Drosophila 80S ribosome. Nature. 2013;497:80–85. doi: 10.1038/nature12104
  229. Hansen T.B., Veno M.T., Jensen T.I., Schaefer A., Damgaard C.K., Kjems J. Argonaute-associated short introns are a novel class of gene regulators. Nat. Commun. 2016;7:11538. doi: 10.1038/ncomms11538
  230. Tenaillon M.I., Hollister J.D., Gaut B.S. A triptych of the evolution of plant transposable elements. Trends Plant Sci. 2010;15:471–478. doi: 10.1016/j.tplants.2010.05.003
  231. Wicker T., Sabot F., Hua-Van A., Bennetzen J.L., Capy P., Chalhoub B., Flavell A., Leroy P., Morgante M., Panaud O. et al. A unified classification system for eukaryotic transposable elements. Nat. Rev. Genet. 2007;8:973–982. doi: 10.1038/nrg2165
  232. Vitte C., Fustier M.A., Alix K., Tenaillon M.I. The bright side of transposons in crop evolution. Brief. Funct. Genomics. 2017;13:276–295. doi: 10.1093/bfgp/elu002
  233. Grandbastien M.A. Activation of plant retrotransposons under stress conditions. Trends Plant Sci. 1998;3:181–187. doi: 10.1016/S1360-1385(98)01232-1
  234. Matzke M.A., Mosher R.A. RNA-directed DNA methylation: an epigenetic pathway of increasing complexity. Nat. Rev. Genet. 2014;15:394–408. doi: 10.1038/nrg3683
  235. Blevins T., Podicheti R., Mishra V., Marasco M., Wang J., Rusch D., Tang H., Pikaard C.S. Identification of Pol IV and RDR2-dependent precursors of 24 nt siRNAs guiding de novo DNA methylation in Arabidopsis. Elife. 2015;4:e09591. doi: 10.7554/eLife.09591
  236. Zhong X., Du J., Hale C.J., Gallego-Bartolome J., Feng S., Vashisht A.A., Chory J., Wohlschlegel J.A., Patel D.J., Jacobsen S.E. et al. Molecular mechanism of action of plant DRM de novo DNA methyltransferases. Cell. 2014;157:1050–1060. doi: 10.1016/j.cell.2014.03.056
  237. Creasey K.M., Zhai J., Borges F., Van Ex F., Regulski M., Meyers B.C., Martienssen R.A. miRNAs trigger widespread epigenetically activated siRNAs from transposons in Arabidopsis. Nature. 2014;508:411–415. doi: 10.1038/nature13069
  238. Watanabe T., Cheng E.C., Zhong M., Lin H. Retrotransposons and pseudogenes regulate mRNAs and lncRNAs via the piRNA pathway in the germline. Genome Res. 2015;25:368–380. doi: 10.1101/gr.180802.114
  239. Wang D., Qu Z., Yang L., Zhang Q., Liu Z.H., Do T., Adelson D.L., Wang Z.-Y., Searle I., Zhu J.-K. Transposable elements (TEs) contribute to stress-related long intergenic noncoding RNAs in plants. Plant J. 2017;90:133–146. doi: 10.1111/tpj.13481
  240. Wang X., Ai G., Zhang C., Cui L., Wang J., Li H., Zhang J., Ye Z. Expression and diversification analysis reveals transposable elements play important roles in the origin of Lycopersicon-specific lncRNAs in tomato. New Phytol. 2015;209:1442–1455. doi: 10.1111/nph.13718
  241. Liu J., Jung C., Xu J., Wang H., Deng S., Bernad L., Arenas-Huertero C., Chua N.-H. Genome-wide analysis uncovers regulation of long intergenic noncoding RNAs in Arabidopsis. Plant Cell. 2012;24:4333–4345. doi: 10.1105/tpc.112.102855
  242. Cao X., Yeo G., Muotri A.R., Kuwabara T., Gage F.H. Noncoding RNAs in the mammalian central nervous system. Annu Rev. Neurosci. 2006;29:77–103. doi: 10.1146/annurev.neuro.29.051605.112839
  243. Mercer T.R., Dinger M.E., Mattick J.S. Long non-coding RNAs: Insights into functions. Nat. Rev. Genet. 2009;10:155–159. doi: 10.1038/nrg2521
  244. Ponting C.P., Oliver P.L., Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136:629–641. doi: 10.1016/j.cell.2009.02.006
  245. Managadze D., Rogozin I.B., Chernikova D., Shabalina S.A., Koonin E.V. Negative correlation between expression level and evolutionary rate of long intergenic noncoding RNAs. Genome Biol. Evol. 2011;3:1390–1404. doi: 10.1093/gbe/evr116
  246. Ponting C.P., Belgard T.G. Transcribed dark matter: meaning or myth? Hum. Mol. Genet. 2010;19:R162–R168. doi: 10.1093/hmg/ddq362
  247. Ponjavic J., Ponting C.P., Lunter G. Functionality or transcriptional noise? Evidence for selection within long noncoding RNAs. Genome Res. 2007;17:556–565. doi: 10.1101/gr.6036807
  248. Pang K.C., Frith M.C., Mattick J.S. Rapid evolution of noncoding RNAs: lack of conservation does not mean lack of function. Trends Genet. 2006;22:1–5. doi: 10.1016/j.tig.2005.10.003
  249. Rinn J.L., Chang H.Y. Genome regulation by long noncoding RNAs. Annu. Rev. Biochem. 2012;81:145–166. doi: 10.1146/annurev-biochem-051410-092902
  250. Wang K.C., Chang H.Y. Molecular mechanisms of long noncoding RNAs. Mol. Cell. 2011;43:904–914. doi: 10.1016/j.molcel.2011.08.018
  251. Guttman M., Amit I., Garber M., French C., Lin M.F., Feldser D., Huarte M., Zuk O., Carey B.W., Cassady J.P. et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 2009;458:223–227. doi: 10.1038/nature07672
  252. Guenther M.G., Levine S.S., Boyer L.A., Jaenisch R., Young R.A. A chromatin landmark and transcription initiation at most promoters in human cells. Cell. 2007;130:77–88. doi: 10.1016/j.cell.2007.05.042
  253. Spitale R.C., Tsai M.C., Chang H.Y. RNA templating the epigenome: Long noncoding RNAs as molecular scaffolds. Epigenetics. 2011;6:539–543. doi: 10.4161/epi.6.5.15221
  254. Good M.C., Zalatan J.G., Lim W.A. Scaffold proteins: hubs for controlling the flow of cellular information. Science. 2011;332:680–686. doi: 10.1126/science
  255. Duret L., Chureau C., Samain S., Weissenbach J., Avner P. The Xist RNA gene evolved in eutherians by pseudogenization of a protein-coding gene. Science. 2006;312:1653–1655. doi: 10.1126/science.1126316
  256. Elisaphenko E.A., Kolesnikov N.N., Shevchenko A.I., Rogozin I.B., Nesterova T.B., Brockdorff N., Zakian S.M. A dual origin of the Xist gene from a protein-coding gene and a set of transposable elements. PLoS ONE. 2008;3:e2521. doi: 10.1371/journal.pone.0002521
  257. Hadjiargyrou M., Delihas N. The intertwining of transposable elements and non-coding RNAs. Int. J. Mol. Sci. 2013;14:13307–13328. doi: 10.3390/ijms140713307
  258. Piriyapongsa J., Jordan I.K. A family of human microRNA genes from miniature inverted-repeat transposable elements. PLoS ONE. 2007;2:e203. doi: 10.1371/journal.pone.0000203
  259. Yuan Z., Sun X., Jiang D., Ding Y., Lu Z., Gong L., Liu H., Xie J. Origin and evolution of a placental-specific microRNA family in the human genome. BMC Evol. Biol. 2010;10:346. doi: 10.1186/1471-2148-10-346
  260. Ahn K., Gim J.-A., Ha H.-S., Han K., Kim H.-S. The novel MER transposon-derived miRNAs in human genome. Gene. 2013;512:422–428. doi: 10.1016/j.gene.2012.08.028
  261. Kannan S., Chernikova D., Rogozin I.B., Poliakov E., Managadze D., Koonin E.V., Milanesi L. Transposable element insertions in long intergenic non-coding RNA genes. Front. Bioeng. Biotechnol. 2015;3:71. doi: 10.3389/fbioe.2015.00071
  262. Kelley D., Rinn J. Transposable elements reveal a stem cell-specific class of long noncoding RNAs. Genome Biol. 2012;13(11):R107. doi: 10.1186/gb-2012-13-11-r107
  263. Cabili M.N., Trapnell C., Goff L., Koziol M., Tazon-Vega B., Regev A., Rinn J.L. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 2011;25:1915–1927. doi: 10.1101/gad.17446611
  264. Ali A., Han K., Liang P. Role of Transposable Elements in Gene Regulation in the Human Genome. Life (Basel). 2021;11(2):118. doi: 10.3390/life11020118
  265. Managadze D., Lobkovsky A.E., Wolf Y.I., Shabalina S.A., Rogozin I.B., Koonin E.V. The Vast, Conserved Mammalian lincRNome. PLoS Comput Biol. 2013;9(2):e1002917. doi: 10.1371/journal.pcbi.1002917
  266. Song Z., Lin J., Li Z., Huang C. The nuclear functions of long noncoding RNAs come into focus. Noncoding RNA Res. 2021;6(2):70–79. doi: 10.1016/j.ncrna.2021.03.002
  267. Braunschweig U., Gueroussov S., Plocik A.M., Graveley B.R., Blencowe B.J. Dynamic integration of splicing within gene regulatory pathways. Cell. 2013;152(6):1252–1269. doi: 10.1016/j.cell.2013.02.034
  268. Wang E.T., Sandberg R., Luo S., Khrebtukova I., Zhang L., Mayr C., Kingsmore S.F., Schroth G.P., Burge C.B. Alternative isoform regulation in human tissue transcriptomes. Nature. 2008;456(7221):470–476. doi: 10.1038/nature07509
  269. Enuka Y., Lauriola M., Feldman M.E., Sas-Chen A., Ulitsky I., Yarden Y. Circular RNAs are long-lived and display only minimal early alterations in response to a growth factor. Nucleic Acids Res. 2016;44(3):1370–1383. doi: 10.1093/nar/gkv1367
  270. Hsu M.T., Coca-Prados M. Electron microscopic evidence for the circular form of RNA in the cytoplasm of eukaryotic cells. Nature. 1979;280(5720):339–340. doi: 10.1038/280339a0
  271. Capel B., Swain A., Nicolis S., Hacker A., Walter M., Koopman P., Goodfellow P., Lovell-Badge R. Circular transcripts of the testis-determining gene Sry in adult mouse testis. Cell. 1993;73(5):1019–1030. doi: 10.1016/0092-8674(93)90279-Y
  272. Cocquerelle C., Daubersies P., Majerus M.A., Kerckaert J.P., Bailleul B. Splicing with inverted order of exons occurs proximal to large introns. EMBO J. 1992;11(3):1095–1098. doi: 10.1002/j.1460-2075.1992.tb05148.x
  273. Nigro J.M., Cho K.R., Fearon E.R., Kern S.E., Ruppert J.M., Oliner J.D., Kinzler K.W., Vogelstein B. Scrambled exons. Cell. 1991;64(3):607–613. doi: 10.1016/0092-8674(91)90244-S
  274. Zaphiropoulos P.G. Circular RNAs from transcripts of the rat cytochrome P450 2C24 gene: correlation with exon skipping. PNAS. 1996;93(13):6536–6541. doi: 10.1073/pnas.93.13.6536
  275. Zaphiropoulos P.G. Exon skipping and circular RNA formation in transcripts of the human cytochrome P-450 2C18 gene in epidermis and of the rat androgen binding protein gene in testis. Mol. Cell Biol. 1997;17(6):2985–2993. doi: 10.1128/MCB.17.6.2985
  276. 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(5):870–885. doi: 10.1016/j.molcel.2015.03.027
  277. 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. doi: 10.1371/journal.pgen.1003777
  278. Jeck W.R., Sorrentino J.A., Wang K., Slevin M.K., Burd C.E., Liu J., Marzluff W.F., Sharpless N.E. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA. 2013;19(2):141–157. doi: 10.1261/rna.035667.112
  279. Salzman J., Gawad C., Wang P.L., Lacayo N., Brown P.O. Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS One. 2012;7(2):e30733. doi: 10.1371/journal.pone.0030733
  280. Guo J.U., Agarwal V., Guo H., Bartel D.P. Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 2014;15(7409). doi: 10.1186/s13059-014-0409-z
  281. Salzman J., Gawad C., Wang P.L., Lacayo N., Brown P.O. Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS ONE. 2012;7:e30733. doi: 10.1371/journal.pone.0030733
  282. Guo J.U., Agarwal V., Guo H., Bartel D.P. Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 2014;15(409). doi: 10.1186/s13059-014-0409-z
  283. Du W.W., Yang W., Liu E., Yang Z., Dhaliwal P., Yang B.B. Foxo3 circular RNA retards cell cycle progression via forming ternary complexes with p21 and CDK2. Nucleic Acids Res. 2016;44:2846–2858. doi: 10.1093/nar/gkw027
  284. Abdelmohsen K., Panda A.C., Munk R., Grammatikakis I., Dudekula D.B., De S., Kim J., Noh J.H., Kim K.M., Martindale J.L., Gorospe M. Identification of HuR target circular RNAs uncovers suppression of PABPN1 translation by CircPABPN1. RNA Biol. 2017;14(3):361–369. doi: 10.1080/15476286.2017.1279788
  285. Zhao R.T., Zhou J., Dong X.L., Bi C.W., Jiang R.C., Dong J.F., Tian Y., Yuan H.J., Zhang J.N. Circular Ribonucleic Acid Expression Alteration in Exosomes from the Brain Extracellular Space after Traumatic Brain Injury in Mice. J. Neurotrauma. 2018;35:2056–2066. doi: 10.1089/neu.2017.5502
  286. Preusser C., Hung L.H., Schneider T., Schreiner S., Hardt M., Moebus A., Santoso S., Bindereif A. Selective release of circRNAs in platelet-derived extracellular vesicles. J. Extracell Vesicles. 2018;7(1):1424473. doi: 10.1080/20013078.2018.1424473
  287. Legnini I., Di Timoteo G., Rossi F., Morlando M., Briganti F., Sthandier O., Fatica A., Santini T., Andronache A., Wade M., et al. Circ-ZNF609 is a circular RNA that can be translated and functions in Myogenesis. Mol. Cell. 2017;66(1):22–37.e9. doi: 10.1016/j.molcel.2017.02.017
  288. Wang Y., Wang Z. Efficient backsplicing produces translatable circular mRNAs. RNA. 2015;21(2):172–179. doi: 10.1261/rna.048272.114
  289. Pamudurti N.R., Bartok O., Jens M., Ashwal-Fluss R., Stottmeister C., Ruhe L., Hanan M., Wyler E., Perez-Hernandez D., Ramberger E. et al. Translation of CircRNAs. Mol. Cell. 2017;66(1):9–21.e7. doi: 10.1016/j.molcel.2017.02.021
  290. Zhang M., Zhao K., Xu X., Yang Y., Yan S., Wei P., Liu H., Xu J., Xiao F., Zhou H. et al. A peptide encoded by circular form of LINC-PINT suppresses oncogenic transcriptional elongation in glioblastoma. Nat. Commun. 2018;9(1):4475. doi: 10.1038/s41467-018-06862-2
  291. Zhang X.O., Wang H.B., Zhang Y., Lu X., Chen L.L., Yang L. Complementary sequence-mediated exon circularization. Cell. 2014;159(1):134–147. doi: 10.1016/j.cell.2014.09.001
  292. 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(1):55–66. doi: 10.1016/j.molcel.2014.08.019
  293. Westholm J.O., Miura P., Olson S., Shenker S., Joseph B., Sanfilippo P., Celniker S.E., Graveley B.R., Lai E.C. Genome-wide analysis of drosophila circular RNAs reveals their structural and sequence properties and age-dependent neural accumulation. Cell Rep. 2014;9(5):1966–1980. doi: 10.1016/j.celrep.2014.10.062
  294. Barrett S.P., Wang P.L., Salzman J. Circular RNA biogenesis can proceed through an exon-containing lariat precursor. Elife. 2015;4:e07540. doi: 10.7554/eLife.07540
  295. Kristensen L.S., Okholm T.L.H., Veno M.T., Kjems J. Circular RNAs are abundantly expressed and upregulated during human epidermal stem cell differentiation. RNA Biol. 2018;15(2):280–291. doi: 10.1080/15476286.2017.1409931
  296. Kramer M.C., Liang D., Tatomer D.C., Gold B., March Z.M., Cherry S., Wilusz J.E. Combinatorial control of Drosophila circular RNA expression by intronic repeats, hnRNPs, and SR proteins. Genes Dev. 2015;29(20):2168–2182. doi: 10.1101/gad.270421.115
  297. Zhang Y., Xue W., Li X., Zhang J., Chen S., Zhang J.L., Yang L., Chen L.L. The biogenesis of nascent circular RNAs. Cell Rep. 2016;15(3):611–624. doi: 10.1016/j.celrep.2016.03.058
  298. Wilusz J.E. A 360 degrees view of circular RNAs: from biogenesis to functions. Wiley Interdiscip. Rev. RNA. 2018;9(4):e1478. doi: 10.1002/wrna.1478
  299. Xiao M.S., Ai Y., Wilusz J.E. Biogenesis and functions of circular RNAs come into focus. Trends Cell Biol. 2020;30(3):226–240. doi: 10.1016/j.tcb.2019.12.004
  300. Yu C.Y., Kuo H.C. The emerging roles and functions of circular RNAs and their generation. J. Biomed. Sci. 2019;26:29. doi: 10.1186/s12929-019-0523-z
  301. Glazar P., Papavasileiou P., Rajewsky N. circBase: a database for circular RNAs. RNA. 2014;20(11):1666–16670. doi: 10.1261/rna.043687.113
  302. Liu Y.C., Li J.R., Sun C.H., Andrews E., Chao R.F., Lin F.M., Weng S.L., Hsu S.D., Huang C.C., Cheng C. et al. CircNet: a database of circular RNAs derived from transcriptome sequencing data. Nucleic Acids Res. 2016;44(D1):D209–D215. doi: 10.1093/nar/gkv940
  303. Ghosal S., Das S., Sen R., Basak P., Chakrabarti J. Circ2Traits: a comprehensive database for circular RNA potentially associated with disease and traits. Front. Genet. 2013;4(283). doi: 10.3389/fgene.2013.00283
  304. Yao D., Zhang L., Zheng M., Sun X., Lu Y., Liu P. Circ2Disease: a manually curated database of experimentally validated circRNAs in human disease. Sci. Rep. 2018;8(1):11018. doi: 10.1038/s41598-018-29360-3
  305. Li S., Li Y., Chen B., Zhao J., Yu S., Tang Y., Zheng Q., Li Y., Wang P., He X., Huang S. exoRBase: a database of circRNA, lncRNA and mRNA in human blood exosomes. Nucleic Acids Res. 2018;46:D106–D112. doi: 10.1093/nar/gkx891
  306. Xia S., Feng J., Chen K., Ma Y., Gong J., Cai F., Jin Y., Gao Y., Xia L., Chang H. et al. CSCD: a database for cancer-specific circular RNAs. Nucleic Acids Res. 2018;46(D1):D925–D929.
  307. Laudadio I., Carissimi C., Fulci V. How RNAi machinery enters the world of telomerase. Cell Cycle. 2019;18:1056–1067. doi: 10.1080/15384101.2019.1609834
  308. Altman S. Ribonuclease P. Philos. Trans. R. Soc. Lond. B. Biol Sci. 2011;366:2936–2941. doi: 10.1098/rstb.2011.0142
  309. Arnold P.R., Wells A.D., Li X.C. Diversity and Emerging Roles of Enhancer RNA in Regulation of Gene Expression and Cell Fate. Frontiers in Cell and Developmental Biology. 2020;7(377). doi: 10.3389/fcell.2019.00377
  310. Bhogireddy S., Mangrauthia S.K., Kumar R., Pandey A.K., Singh S., Jain A., Budak H., Varshney R.K., Kudapa H. Regulatory non-coding RNAs: a new frontier in regulation of plant biology. Funct. Integr. Genomics. 2021;21:313–330. doi: 10.1007/s10142-021-00787-8
  311. Song L., Fang Y., Chen L., Wang J., Chen X. Role of non-coding RNAs in plant immunity. Plant Commun. 2021;2(100180). doi: 10.1016/j.xplc.2021.100180
  312. Wu N., Yang B.B. The Biological Functions of Non-coding RNAs: From a Line to a Circle. Discoveries (Craiova). 2015;3:e48. doi: 10.15190/d.2015.40
  313. Cerutti H., Casas-Mollano J.A. On the origin and functions of RNA-mediated silencing: from protists to man. Curr. Genet. 2006;50:81–99. doi: 10.1007/s00294-006-0078-x
  314. Grigoriev A. Transfer RNA and Origins of RNA Interference. Front Mol Biosci. 2021;8(708984). doi: 10.3389/fmolb.2021.708984
  315. Mohammed J., Flynt A.S., Panzarino A.M., Mondal M.M.H., DeCruz M., Siepel A., Lai E.C. Deep experimental profiling of microRNA diversity, deployment, and evolution across the Drosophila genus. Genome Res. 2018;28:52–65. doi: 10.1101/gr.226068.117
  316. Gilbert W. Origin of life: The RNA world. Nature. 1986;319:618. doi: 10.1038/319618a0
  317. Crick F.H. The origin of the genetic code. J. Mol. Biol. 1968;38:367–379. doi: 10.1016/0022-2836(68)90392-6
  318. Orgel L.E Evolution of the genetic apparatus. J. Mol. Biol. 1968;38:381–393. doi: 10.1016/0022-2836(68)90393-8
  319. Woese C.R. The genetic code: The molecular basis for genetic expression. Harper & Row, 1967. 186 p.
  320. Eddy S.R. Computational genomics of noncoding RNA genes. Cell. 2002;109:137–140. doi: 10.1016/S0092-8674(02)00727-4
  321. RNA Worlds: From Life's Origins to Diversity in Gene Regulation. Eds.: Atkins J.F., Gesteland R.F., Cech T.R. Cold Spring Harbor Laboratory Press, 2011. 366 p.
  322. Koonin E.V. The logic of chance. The nature and origin of biological evolution. Pearson Education, Inc.; 2012. ISBN: 0132542498
  323. Bokov K., Steinberg S.V. A hierarchical model for evolution of 23S ribosomal RNA. Nature. 2009;457:977–980. doi: 10.1038/nature07749
  324. Skoblikow N.E., Zimin А.А. A search for relict ribonucleotide and amino acid sequences that played a key role in the development of the ribosome and modern protein diversity. Math. Biol. Bioinf. 2015;10:116–130. doi: 10.17537/2015.10.116
  325. Nazipova N.N., Isaev E.A., Kornilov V.V., Pervukhin D.V., Morozova A.A., Gorbunov A.A., Ustinin M.N. Big Data in Bioinformatics. Math. Biol. Bioinf. 2018;13:t1–t16. doi: 10.17537/2018.13.t1

 

Содержание Оригинальная статья
Мат. биол. и биоинф.
2021;16(2):256-298
doi: 10.17537/2021.16.256
опубликована на рус. яз.

Аннотация (рус.)
Аннотация (англ.)
Полный текст (рус., pdf)
Список литературы

 

  Copyright ИМПБ РАН © 2005-2024