#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

SmD1 Modulates the miRNA Pathway Independently of Its Pre-mRNA Splicing Function


microRNAs (miRNAs) are a class of small regulatory RNAs that fine-tune gene expression by reducing protein output from their target messenger RNAs and are implicated in myriad physiological and pathological processes. miRNAs are generated from long primary transcripts via sequential actions of the Drosha/Pasha and Dicer ribonucleases. Mature miRNAs are incorporated into the miRISC effector complexes that contain AGO family member proteins and serve as specificity determinants to guide miRISCs to their target RNAs. Previous studies suggested that select proteins implicated in the processing of messenger RNAs are required for the miRNA production/function, but the underlying molecular mechanism is not well understood. Here we show that SmD1, an essential protein implicated in the processing of messenger RNAs, directly interacts with both Pasha and primary miRNA transcripts and is required for optimal miRNA production. Furthermore, SmD1 associates with multiple components of the miRNA effector machinery and is required for miRNA function. Finally, our analysis reveals that defects in the miRNA pathway can be uncoupled from those in messenger RNA processing, and that the miRNA biogenesis and messenger RNA processing machineries are physically and functionally distinct entities. Our data thus suggests that SmD1 modulates the miRNA pathway independent of its role in messenger RNA processing.


Vyšlo v časopise: SmD1 Modulates the miRNA Pathway Independently of Its Pre-mRNA Splicing Function. PLoS Genet 11(8): e32767. doi:10.1371/journal.pgen.1005475
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005475

Souhrn

microRNAs (miRNAs) are a class of small regulatory RNAs that fine-tune gene expression by reducing protein output from their target messenger RNAs and are implicated in myriad physiological and pathological processes. miRNAs are generated from long primary transcripts via sequential actions of the Drosha/Pasha and Dicer ribonucleases. Mature miRNAs are incorporated into the miRISC effector complexes that contain AGO family member proteins and serve as specificity determinants to guide miRISCs to their target RNAs. Previous studies suggested that select proteins implicated in the processing of messenger RNAs are required for the miRNA production/function, but the underlying molecular mechanism is not well understood. Here we show that SmD1, an essential protein implicated in the processing of messenger RNAs, directly interacts with both Pasha and primary miRNA transcripts and is required for optimal miRNA production. Furthermore, SmD1 associates with multiple components of the miRNA effector machinery and is required for miRNA function. Finally, our analysis reveals that defects in the miRNA pathway can be uncoupled from those in messenger RNA processing, and that the miRNA biogenesis and messenger RNA processing machineries are physically and functionally distinct entities. Our data thus suggests that SmD1 modulates the miRNA pathway independent of its role in messenger RNA processing.


Zdroje

1. Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, et al. (2000) The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 403: 901–906. 10706289

2. Lee RC, Feinbaum RL, Ambros V (1993) The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75: 843–854. 8252621

3. Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, et al. (2004) The Microprocessor complex mediates the genesis of microRNAs. Nature 432: 235–240. 15531877

4. Lee Y, Ahn C, Han J, Choi H, Kim J, et al. (2003) The nuclear RNase III Drosha initiates microRNA processing. Nature 425: 415–419. 14508493

5. Denli AM, Tops BB, Plasterk RH, Ketting RF, Hannon GJ (2004) Processing of primary microRNAs by the Microprocessor complex. Nature 432: 231–235. 15531879

6. Saito K, Ishizuka A, Siomi H, Siomi MC (2005) Processing of pre-microRNAs by the Dicer-1-Loquacious complex in Drosophila cells. PLoS Biol 3: e235. 15918769

7. Hutvagner G, McLachlan J, Pasquinelli AE, Balint E, Tuschl T, et al. (2001) A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science 293: 834–838. 11452083

8. Forstemann K, Tomari Y, Du T, Vagin VV, Denli AM, et al. (2005) Normal microRNA maturation and germ-line stem cell maintenance requires Loquacious, a double-stranded RNA-binding domain protein. PLoS Biol 3: e236. 15918770

9. Jiang F, Ye X, Liu X, Fincher L, McKearin D, et al. (2005) Dicer-1 and R3D1-L catalyze microRNA maturation in Drosophila. Genes Dev 19: 1674–1679. 15985611

10. Lund E, Guttinger S, Calado A, Dahlberg JE, Kutay U (2004) Nuclear export of microRNA precursors. Science 303: 95–98. 14631048

11. Yi R, Qin Y, Macara IG, Cullen BR (2003) Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev 17: 3011–3016. 14681208

12. Bohnsack MT, Czaplinski K, Gorlich D (2004) Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA 10: 185–191. 14730017

13. Guo H, Ingolia NT, Weissman JS, Bartel DP (2010) Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature 466: 835–840. doi: 10.1038/nature09267 20703300

14. Pillai RS, Bhattacharyya SN, Artus CG, Zoller T, Cougot N, et al. (2005) Inhibition of translational initiation by Let-7 MicroRNA in human cells. Science 309: 1573–1576. 16081698

15. Fabian MR, Mathonnet G, Sundermeier T, Mathys H, Zipprich JT, et al. (2009) Mammalian miRNA RISC recruits CAF1 and PABP to affect PABP-dependent deadenylation. Mol Cell 35: 868–880. doi: 10.1016/j.molcel.2009.08.004 19716330

16. Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB (2003) Prediction of mammalian microRNA targets. Cell 115: 787–798. 14697198

17. Lai EC, Tam B, Rubin GM (2005) Pervasive regulation of Drosophila Notch target genes by GY-box-, Brd-box-, and K-box-class microRNAs. Genes Dev 19: 1067–1080. 15833912

18. Lai EC, Burks C, Posakony JW (1998) The K box, a conserved 3' UTR sequence motif, negatively regulates accumulation of enhancer of split complex transcripts. Development 125: 4077–4088. 9735368

19. Taliaferro JM, Aspden JL, Bradley T, Marwha D, Blanchette M, et al. (2013) Two new and distinct roles for Drosophila Argonaute-2 in the nucleus: alternative pre-mRNA splicing and transcriptional repression. Genes Dev 27: 378–389. doi: 10.1101/gad.210708.112 23392611

20. Ameyar-Zazoua M, Rachez C, Souidi M, Robin P, Fritsch L, et al. (2012) Argonaute proteins couple chromatin silencing to alternative splicing. Nat Struct Mol Biol 19: 998–1004. doi: 10.1038/nsmb.2373 22961379

21. Bayne EH, Portoso M, Kagansky A, Kos-Braun IC, Urano T, et al. (2008) Splicing factors facilitate RNAi-directed silencing in fission yeast. Science 322: 602–606. doi: 10.1126/science.1164029 18948543

22. Herr AJ, Molnar A, Jones A, Baulcombe DC (2006) Defective RNA processing enhances RNA silencing and influences flowering of Arabidopsis. Proc Natl Acad Sci U S A 103: 14994–15001. 17008405

23. Guil S, Caceres JF (2007) The multifunctional RNA-binding protein hnRNP A1 is required for processing of miR-18a. Nat Struct Mol Biol 14: 591–596. 17558416

24. Michlewski G, Caceres JF (2010) Antagonistic role of hnRNP A1 and KSRP in the regulation of let-7a biogenesis. Nat Struct Mol Biol 17: 1011–1018. doi: 10.1038/nsmb.1874 20639884

25. Ruggiero T, Trabucchi M, De Santa F, Zupo S, Harfe BD, et al. (2009) LPS induces KH-type splicing regulatory protein-dependent processing of microRNA-155 precursors in macrophages. FASEB J 23: 2898–2908. doi: 10.1096/fj.09-131342 19423639

26. Trabucchi M, Briata P, Garcia-Mayoral M, Haase AD, Filipowicz W, et al. (2009) The RNA-binding protein KSRP promotes the biogenesis of a subset of microRNAs. Nature 459: 1010–1014. doi: 10.1038/nature08025 19458619

27. Zhou R, Hotta I, Denli AM, Hong P, Perrimon N, et al. (2008) Comparative analysis of argonaute-dependent small RNA pathways in Drosophila. Mol Cell 32: 592–599. doi: 10.1016/j.molcel.2008.10.018 19026789

28. Kim JK, Gabel HW, Kamath RS, Tewari M, Pasquinelli A, et al. (2005) Functional genomic analysis of RNA interference in C. elegans. Science 308: 1164–1167. 15790806

29. Parry DH, Xu J, Ruvkun G (2007) A whole-genome RNAi Screen for C. elegans miRNA pathway genes. Curr Biol 17: 2013–2022. 18023351

30. Tabach Y, Billi AC, Hayes GD, Newman MA, Zuk O, et al. (2013) Identification of small RNA pathway genes using patterns of phylogenetic conservation and divergence. Nature 493: 694–698. doi: 10.1038/nature11779 23364702

31. Kambach C, Walke S, Young R, Avis JM, de la Fortelle E, et al. (1999) Crystal structures of two Sm protein complexes and their implications for the assembly of the spliceosomal snRNPs. Cell 96: 375–387. 10025403

32. Mount SM, Salz HK (2000) Pre-messenger RNA processing factors in the Drosophila genome. J Cell Biol 150: F37–44. 10908584

33. Xiong XP, Kurthkoti K, Chang KY, Lichinchi G, De N, et al. (2013) Core small nuclear ribonucleoprotein particle splicing factor SmD1 modulates RNA interference in Drosophila. Proc Natl Acad Sci U S A 110: 16520–16525. doi: 10.1073/pnas.1315803110 24067655

34. Brennecke J, Hipfner DR, Stark A, Russell RB, Cohen SM (2003) bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell 113: 25–36. 12679032

35. Stark A, Brennecke J, Russell RB, Cohen SM (2003) Identification of Drosophila MicroRNA targets. PLoS Biol 1: E60. 14691535

36. Sempere LF, Sokol NS, Dubrovsky EB, Berger EM, Ambros V (2003) Temporal regulation of microRNA expression in Drosophila melanogaster mediated by hormonal signals and broad-Complex gene activity. Dev Biol 259: 9–18. 12812784

37. Kadener S, Rodriguez J, Abruzzi KC, Khodor YL, Sugino K, et al. (2009) Genome-wide identification of targets of the drosha-pasha/DGCR8 complex. RNA 15: 537–545. doi: 10.1261/rna.1319309 19223442

38. Han J, Pedersen JS, Kwon SC, Belair CD, Kim YK, et al. (2009) Posttranscriptional crossregulation between Drosha and DGCR8. Cell 136: 75–84. doi: 10.1016/j.cell.2008.10.053 19135890

39. Yeom KH, Lee Y, Han J, Suh MR, Kim VN (2006) Characterization of DGCR8/Pasha, the essential cofactor for Drosha in primary miRNA processing. Nucleic Acids Res 34: 4622–4629. 16963499

40. Nguyen TA, Jo MH, Choi YG, Park J, Kwon SC, et al. (2015) Functional Anatomy of the Human Microprocessor. Cell 161: 1374–1387. doi: 10.1016/j.cell.2015.05.010 26027739

41. Hafner M, Landthaler M, Burger L, Khorshid M, Hausser J, et al. (2010) Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell 141: 129–141. doi: 10.1016/j.cell.2010.03.009 20371350

42. Martin G, Gruber AR, Keller W, Zavolan M (2012) Genome-wide analysis of pre-mRNA 3' end processing reveals a decisive role of human cleavage factor I in the regulation of 3' UTR length. Cell Rep 1: 753–763. doi: 10.1016/j.celrep.2012.05.003 22813749

43. Morlando M, Dini Modigliani S, Torrelli G, Rosa A, Di Carlo V, et al. (2012) FUS stimulates microRNA biogenesis by facilitating co-transcriptional Drosha recruitment. EMBO J 31: 4502–4510. doi: 10.1038/emboj.2012.319 23232809

44. Lehmeier T, Raker V, Hermann H, Luhrmann R (1994) cDNA cloning of the Sm proteins D2 and D3 from human small nuclear ribonucleoproteins: evidence for a direct D1-D2 interaction. Proc Natl Acad Sci U S A 91: 12317–12321. 7527560

45. Duchaine TF, Wohlschlegel JA, Kennedy S, Bei Y, Conte D Jr., et al. (2006) Functional proteomics reveals the biochemical niche of C. elegans DCR-1 in multiple small-RNA-mediated pathways. Cell 124: 343–354. 16439208

46. Czech B, Malone CD, Zhou R, Stark A, Schlingeheyde C, et al. (2008) An endogenous small interfering RNA pathway in Drosophila. Nature 453: 798–802. doi: 10.1038/nature07007 18463631

47. Miyoshi K, Okada TN, Siomi H, Siomi MC (2009) Characterization of the miRNA-RISC loading complex and miRNA-RISC formed in the Drosophila miRNA pathway. RNA 15: 1282–1291. doi: 10.1261/rna.1541209 19451544

48. Zhou R, Czech B, Brennecke J, Sachidanandam R, Wohlschlegel JA, et al. (2009) Processing of Drosophila endo-siRNAs depends on a specific Loquacious isoform. RNA 15: 1886–1895. doi: 10.1261/rna.1611309 19635780

49. Ishizuka A, Saito K, Siomi MC, Siomi H (2006) In vitro precursor microRNA processing assays using Drosophila Schneider-2 cell lysates. Methods Mol Biol 342: 277–286. 16957382

50. Nayak A, Andino R (2011) Slicer activity in Drosophila melanogaster S2 extract. Methods Mol Biol 721: 231–244. doi: 10.1007/978-1-61779-037-9_14 21431689

51. Dodt M, Roehr JT, Ahmed R, Dieterich C (2012) FLEXBAR-Flexible Barcode and Adapter Processing for Next-Generation Sequencing Platforms. Biology (Basel) 1: 895–905.

52. Langmead B, Trapnell C, Pop M, Salzberg SL (2009) Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 10: R25. doi: 10.1186/gb-2009-10-3-r25 19261174

53. Corcoran DL, Georgiev S, Mukherjee N, Gottwein E, Skalsky RL, et al. (2011) PARalyzer: definition of RNA binding sites from PAR-CLIP short-read sequence data. Genome Biol 12: R79. doi: 10.1186/gb-2011-12-8-r79 21851591

54. Anders S, Pyl PT, Huber W (2015) HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31: 166–169. doi: 10.1093/bioinformatics/btu638 25260700

55. Brand AH, Perrimon N (1993) Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Development 118: 401–415. 8223268

56. Lo PC, Skeath JB, Gajewski K, Schulz RA, Frasch M (2002) Homeotic genes autonomously specify the anteroposterior subdivision of the Drosophila dorsal vessel into aorta and heart. Dev Biol 251: 307–319. 12435360

57. Fink M, Callol-Massot C, Chu A, Ruiz-Lozano P, Izpisua Belmonte JC, et al. (2009) A new method for detection and quantification of heartbeat parameters in Drosophila, zebrafish, and embryonic mouse hearts. Biotechniques 46: 101–113. doi: 10.2144/000113078 19317655

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 8
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#