#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

The Intronic Long Noncoding RNA Recruits PRC2 to the Promoter, Reducing the Expression of and Increasing Cell Proliferation


The down-regulation of the tumor-suppressor gene RASSF1A has been shown to increase cell proliferation in several tumors. RASSF1A expression is regulated through epigenetic events involving the polycomb repressive complex 2 (PRC2); however, the molecular mechanisms modulating the recruitment of this epigenetic modifier to the RASSF1 locus remain largely unknown. Here, we identify and characterize ANRASSF1, an endogenous unspliced long noncoding RNA (lncRNA) that is transcribed from the opposite strand on the RASSF1 gene locus in several cell lines and tissues and binds PRC2. ANRASSF1 is transcribed through RNA polymerase II and is 5′-capped and polyadenylated; it exhibits nuclear localization and has a shorter half-life compared with other lncRNAs that bind PRC2. ANRASSF1 endogenous expression is higher in breast and prostate tumor cell lines compared with non-tumor, and an opposite pattern is observed for RASSF1A. ANRASSF1 ectopic overexpression reduces RASSF1A abundance and increases the proliferation of HeLa cells, whereas ANRASSF1 silencing causes the opposite effects. These changes in ANRASSF1 levels do not affect the RASSF1C isoform abundance. ANRASSF1 overexpression causes a marked increase in both PRC2 occupancy and histone H3K27me3 repressive marks, specifically at the RASSF1A promoter region. No effect of ANRASSF1 overexpression was detected on PRC2 occupancy and histone H3K27me3 at the promoter regions of RASSF1C and the four other neighboring genes, including two well-characterized tumor suppressor genes. Additionally, we demonstrated that ANRASSF1 forms an RNA/DNA hybrid and recruits PRC2 to the RASSF1A promoter. Together, these results demonstrate a novel mechanism of epigenetic repression of the RASSF1A tumor suppressor gene involving antisense unspliced lncRNA, in which ANRASSF1 selectively represses the expression of the RASSF1 isoform overlapping the antisense transcript in a location-specific manner. In a broader perspective, our findings suggest that other non-characterized unspliced intronic lncRNAs transcribed in the human genome might contribute to a location-specific epigenetic modulation of genes.


Vyšlo v časopise: The Intronic Long Noncoding RNA Recruits PRC2 to the Promoter, Reducing the Expression of and Increasing Cell Proliferation. PLoS Genet 9(8): e32767. doi:10.1371/journal.pgen.1003705
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003705

Souhrn

The down-regulation of the tumor-suppressor gene RASSF1A has been shown to increase cell proliferation in several tumors. RASSF1A expression is regulated through epigenetic events involving the polycomb repressive complex 2 (PRC2); however, the molecular mechanisms modulating the recruitment of this epigenetic modifier to the RASSF1 locus remain largely unknown. Here, we identify and characterize ANRASSF1, an endogenous unspliced long noncoding RNA (lncRNA) that is transcribed from the opposite strand on the RASSF1 gene locus in several cell lines and tissues and binds PRC2. ANRASSF1 is transcribed through RNA polymerase II and is 5′-capped and polyadenylated; it exhibits nuclear localization and has a shorter half-life compared with other lncRNAs that bind PRC2. ANRASSF1 endogenous expression is higher in breast and prostate tumor cell lines compared with non-tumor, and an opposite pattern is observed for RASSF1A. ANRASSF1 ectopic overexpression reduces RASSF1A abundance and increases the proliferation of HeLa cells, whereas ANRASSF1 silencing causes the opposite effects. These changes in ANRASSF1 levels do not affect the RASSF1C isoform abundance. ANRASSF1 overexpression causes a marked increase in both PRC2 occupancy and histone H3K27me3 repressive marks, specifically at the RASSF1A promoter region. No effect of ANRASSF1 overexpression was detected on PRC2 occupancy and histone H3K27me3 at the promoter regions of RASSF1C and the four other neighboring genes, including two well-characterized tumor suppressor genes. Additionally, we demonstrated that ANRASSF1 forms an RNA/DNA hybrid and recruits PRC2 to the RASSF1A promoter. Together, these results demonstrate a novel mechanism of epigenetic repression of the RASSF1A tumor suppressor gene involving antisense unspliced lncRNA, in which ANRASSF1 selectively represses the expression of the RASSF1 isoform overlapping the antisense transcript in a location-specific manner. In a broader perspective, our findings suggest that other non-characterized unspliced intronic lncRNAs transcribed in the human genome might contribute to a location-specific epigenetic modulation of genes.


Zdroje

1. DonningerH, VosMD, ClarkGJ (2007) The RASSF1A tumor suppressor. J Cell Sci 120: 3163–3172.

2. DammannR, SchagdarsurenginU, StrunnikovaM, RastetterM, SeidelC, et al. (2003) Epigenetic inactivation of the Ras-association domain family 1 (RASSF1A) gene and its function in human carcinogenesis. Histol Histopathol 18: 665–677.

3. DammannR, LiC, YoonJH, ChinPL, BatesS, et al. (2000) Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nat Genet 25: 315–319.

4. AgathanggelouA, CooperWN, LatifF (2005) Role of the Ras-association domain family 1 tumor suppressor gene in human cancers. Cancer Res 65: 3497–3508.

5. AminKS, BanerjeePP (2012) The cellular functions of RASSF1A and its inactivation in prostate cancer. J Carcinog 11: 3.

6. AvruchJ, XavierR, BardeesyN, ZhangXF, PraskovaM, et al. (2009) Rassf family of tumor suppressor polypeptides. J Biol Chem 284: 11001–11005.

7. FernandesMS, CarneiroF, OliveiraC, SerucaR (2012) Colorectal cancer and RASSF family - A special emphasis on RASSF1A. Int J Cancer 132 (2) 251–8 doi: 10.1002/ijc.27696

8. JiangY, CuiL, ChenWD, ShenSH, DingLD (2012) The prognostic role of RASSF1A promoter methylation in breast cancer: a meta-analysis of published data. PLoS One 7: e36780.

9. HoqueMO, BraitM, RosenbaumE, PoetaML, PalP, et al. (2010) Genetic and epigenetic analysis of erbB signaling pathway genes in lung cancer. J Thorac Oncol 5: 1887–1893.

10. PalakurthyRK, WajapeyeeN, SantraMK, GazinC, LinL, et al. (2009) Epigenetic silencing of the RASSF1A tumor suppressor gene through HOXB3-mediated induction of DNMT3B expression. Mol Cell 36: 219–230.

11. GoodliffeJM, WieschausE, ColeMD (2005) Polycomb mediates Myc autorepression and its transcriptional control of many loci in Drosophila. Genes Dev 19: 2941–2946.

12. BirneyE, StamatoyannopoulosJA, DuttaA, GuigoR, GingerasTR, et al. (2007) Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 447: 799–816.

13. KapranovP, ChengJ, DikeS, NixDA, DuttaguptaR, et al. (2007) RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science 316: 1484–1488.

14. WangX, SongX, GlassCK, RosenfeldMG (2011) The long arm of long noncoding RNAs: roles as sensors regulating gene transcriptional programs. Cold Spring Harb Perspect Biol 3: a003756.

15. KhalilAM, RinnJL (2011) RNA-protein interactions in human health and disease. Semin Cell Dev Biol 22: 359–365.

16. MattickJS (2012) RNA driving the epigenetic bus. EMBO J 31: 515–516.

17. GuttmanM, DonagheyJ, CareyBW, GarberM, GrenierJK, et al. (2011) lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature 477: 295–300.

18. KhalilAM, GuttmanM, HuarteM, GarberM, RajA, et al. (2009) Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci U S A 106: 11667–11672.

19. RinnJL, KerteszM, WangJK, SquazzoSL, XuX, et al. (2007) Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 129: 1311–1323.

20. GuilS, SolerM, PortelaA, CarrereJ, FonallerasE, et al. (2012) Intronic RNAs mediate EZH2 regulation of epigenetic targets. Nat Struct Mol Biol 19: 664–670.

21. ZhaoJ, OhsumiTK, KungJT, OgawaY, GrauDJ, et al. (2010) Genome-wide identification of polycomb-associated RNAs by RIP-seq. Mol Cell 40: 939–953.

22. NakayaHI, AmaralPP, LouroR, LopesA, FachelAA, et al. (2007) Genome mapping and expression analyses of human intronic noncoding RNAs reveal tissue-specific patterns and enrichment in genes related to regulation of transcription. Genome Biol 8: R43.

23. DjebaliS, DavisCA, MerkelA, DobinA, LassmannT, et al. (2012) Landscape of transcription in human cells. Nature 489: 101–108.

24. ColganDF, ManleyJL (1997) Mechanism and regulation of mRNA polyadenylation. Genes Dev 11: 2755–2766.

25. KongL, ZhangY, YeZQ, LiuXQ, ZhaoSQ, et al. (2007) CPC: assess the protein-coding potential of transcripts using sequence features and support vector machine. Nucleic Acids Res 35: W345–349.

26. GiovinazziS, LindsayCR, MorozovVM, Escobar-CabreraE, SummersMK, et al. (2012) Regulation of mitosis and taxane response by Daxx and Rassf1. Oncogene 31: 13–26.

27. JiP, DiederichsS, WangW, BoingS, MetzgerR, et al. (2003) MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene 22: 8031–8041.

28. DavuluriRV, GrosseI, ZhangMQ (2001) Computational identification of promoters and first exons in the human genome. Nat Genet 29: 412–417.

29. CalipelA, AbonnetV, NicoleO, MascarelliF, CouplandSE, et al. (2011) Status of RASSF1A in uveal melanocytes and melanoma cells. Mol Cancer Res 9: 1187–1198.

30. MatallanasD, RomanoD, YeeK, MeisslK, KucerovaL, et al. (2007) RASSF1A elicits apoptosis through an MST2 pathway directing proapoptotic transcription by the p73 tumor suppressor protein. Mol Cell 27: 962–975.

31. ZhangXD, GillespieSK, HerseyP (2004) Staurosporine induces apoptosis of melanoma by both caspase-dependent and -independent apoptotic pathways. Molecular Cancer Therapeutics 3: 187–197.

32. UnoF, SasakiJ, NishizakiM, CarboniG, XuK, et al. (2004) Myristoylation of the fus1 protein is required for tumor suppression in human lung cancer cells. Cancer Res 64: 2969–2976.

33. KurataA, KatayamaR, WatanabeT, TsuruoT, FujitaN (2008) TUSC4/NPRL2, a novel PDK1-interacting protein, inhibits PDK1 tyrosine phosphorylation and its downstream signaling. Cancer Sci 99: 1827–1834.

34. CaoR, ZhangY (2004) SUZ12 is required for both the histone methyltransferase activity and the silencing function of the EED-EZH2 complex. Mol Cell 15: 57–67.

35. WangX, AraiS, SongX, ReichartD, DuK, et al. (2008) Induced ncRNAs allosterically modify RNA-binding proteins in cis to inhibit transcription. Nature 454: 126–130.

36. BertaniS, SauerS, BolotinE, SauerF (2011) The noncoding RNA Mistral activates Hoxa6 and Hoxa7 expression and stem cell differentiation by recruiting MLL1 to chromatin. Mol Cell 43: 1040–1046.

37. GuttmanM, AmitI, GarberM, FrenchC, LinMF, et al. (2009) Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 458: 223–227.

38. RedrupL, BrancoMR, PerdeauxER, KruegerC, LewisA, et al. (2009) The long noncoding RNA Kcnq1ot1 organises a lineage-specific nuclear domain for epigenetic gene silencing. Development 136: 525–530.

39. RinnJL, ChangHY (2012) Genome Regulation by Long Noncoding RNAs. Annu Rev Biochem 81: 145–166.

40. PandeyRR, MondalT, MohammadF, EnrothS, RedrupL, et al. (2008) Kcnq1ot1 antisense noncoding RNA mediates lineage-specific transcriptional silencing through chromatin-level regulation. Mol Cell 32: 232–246.

41. EngelhardtJ, StadlerPF (2012) Hidden treasures in unspliced EST data. Theory Biosci 131: 49–57.

42. MondalT, RasmussenM, PandeyGK, IsakssonA, KanduriC (2010) Characterization of the RNA content of chromatin. Genome Res 20: 899–907.

43. LouroR, SmirnovaAS, Verjovski-AlmeidaS (2009) Long intronic noncoding RNA transcription: expression noise or expression choice? Genomics 93: 291–298.

44. WapinskiO, ChangHY (2011) Long noncoding RNAs and human disease. Trends Cell Biol 21: 354–361.

45. TaftRJ, PangKC, MercerTR, DingerM, MattickJS (2010) Non-coding RNAs: regulators of disease. J Pathol 220: 126–139.

46. EstellerM (2011) Non-coding RNAs in human disease. Nat Rev Genet 12: 861–874.

47. SpizzoR, AlmeidaMI, ColombattiA, CalinGA (2012) Long non-coding RNAs and cancer: a new frontier of translational research? Oncogene 31: 4577–4587.

48. PrensnerJR, ChinnaiyanAM (2011) The emergence of lncRNAs in cancer biology. Cancer Discov 1: 391–407.

49. ReisEM, Verjovski-AlmeidaS (2012) Perspectives of Long Non-Coding RNAs in Cancer Diagnostics. Front Genet 3: 32.

50. GutschnerT, DiederichsS (2012) The Hallmarks of Cancer: A long non-coding RNA point of view. RNA Biol 9: 703–719.

51. PfafflMW (2001) A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 29: e45.

52. TrapnellC, PachterL, SalzbergSL (2009) TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25: 1105–1111.

53. TrapnellC, WilliamsBA, PerteaG, MortazaviA, KwanG, et al. (2010) Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol 28: 511–515.

54. CappellenD, SchlangeT, BauerM, MaurerF, HynesNE (2007) Novel c-MYC target genes mediate differential effects on cell proliferation and migration. EMBO Rep 8: 70–76.

55. ByunJS, WongMM, CuiW, IdelmanG, LiQ, et al. (2009) Dynamic bookmarking of primary response genes by p300 and RNA polymerase II complexes. Proc Natl Acad Sci U S A 106: 19286–19291.

56. Lewis-WambiJS, CunliffeHE, KimHR, WillisAL, JordanVC (2008) Overexpression of CEACAM6 promotes migration and invasion of oestrogen-deprived breast cancer cells. Eur J Cancer 44: 1770–1779.

57. PellegriniM, ChengJC, VoutilaJ, JudelsonD, TaylorJ, et al. (2008) Expression profile of CREB knockdown in myeloid leukemia cells. BMC Cancer 8: 264.

58. IrizarryRA, Ladd-AcostaC, WenB, WuZ, MontanoC, et al. (2009) The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet 41: 178–186.

59. MoraesMC, de AndradeAQ, CarvalhoH, GuechevaT, AgnolettoMH, et al. (2012) Both XPA and DNA polymerase eta are necessary for the repair of doxorubicin-induced DNA lesions. Cancer Lett 314: 108–118.

60. Sambrook J, Russell DW (2001) Molecular cloning : a laboratory manual. Cold Spring Harbor, N.Y.: Cold Spring Harbor Laboratory Press.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 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#