#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

ALS-Associated Mutations Result in Compromised Alternative Splicing and Autoregulation


The gene encoding a DNA/RNA binding protein FUS/TLS is frequently mutated in amyotrophic lateral sclerosis (ALS). Mutations commonly affect its carboxy-terminal nuclear localization signal, resulting in varying deficiencies of FUS nuclear localization and abnormal cytoplasmic accumulation. Increasing evidence suggests deficiencies in FUS nuclear function may contribute to neuron degeneration. Here we report a novel FUS autoregulatory mechanism and its deficiency in ALS-associated mutants. Using FUS CLIP-seq, we identified significant FUS binding to a highly conserved region of exon 7 and the flanking introns of its own pre-mRNAs. We demonstrated that FUS is a repressor of exon 7 splicing and that the exon 7-skipped splice variant is subject to nonsense-mediated decay (NMD). Overexpression of FUS led to the repression of exon 7 splicing and a reduction of endogenous FUS protein. Conversely, the repression of exon 7 was reduced by knockdown of FUS protein, and moreover, it was rescued by expression of EGFP-FUS. This dynamic regulation of alternative splicing describes a novel mechanism of FUS autoregulation. Given that ALS-associated FUS mutants are deficient in nuclear localization, we examined whether cells expressing these mutants would be deficient in repressing exon 7 splicing. We showed that FUS harbouring R521G, R522G or ΔExon15 mutation (minor, moderate or severe cytoplasmic localization, respectively) directly correlated with respectively increasing deficiencies in both exon 7 repression and autoregulation of its own protein levels. These data suggest that compromised FUS autoregulation can directly exacerbate the pathogenic accumulation of cytoplasmic FUS protein in ALS. We showed that exon 7 skipping can be induced by antisense oligonucleotides targeting its flanking splice sites, indicating the potential to alleviate abnormal cytoplasmic FUS accumulation in ALS. Taken together, FUS autoregulation by alternative splicing provides insight into a molecular mechanism by which FUS-regulated pre-mRNA processing can impact a significant number of targets important to neurodegeneration.


Vyšlo v časopise: ALS-Associated Mutations Result in Compromised Alternative Splicing and Autoregulation. PLoS Genet 9(10): e32767. doi:10.1371/journal.pgen.1003895
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003895

Souhrn

The gene encoding a DNA/RNA binding protein FUS/TLS is frequently mutated in amyotrophic lateral sclerosis (ALS). Mutations commonly affect its carboxy-terminal nuclear localization signal, resulting in varying deficiencies of FUS nuclear localization and abnormal cytoplasmic accumulation. Increasing evidence suggests deficiencies in FUS nuclear function may contribute to neuron degeneration. Here we report a novel FUS autoregulatory mechanism and its deficiency in ALS-associated mutants. Using FUS CLIP-seq, we identified significant FUS binding to a highly conserved region of exon 7 and the flanking introns of its own pre-mRNAs. We demonstrated that FUS is a repressor of exon 7 splicing and that the exon 7-skipped splice variant is subject to nonsense-mediated decay (NMD). Overexpression of FUS led to the repression of exon 7 splicing and a reduction of endogenous FUS protein. Conversely, the repression of exon 7 was reduced by knockdown of FUS protein, and moreover, it was rescued by expression of EGFP-FUS. This dynamic regulation of alternative splicing describes a novel mechanism of FUS autoregulation. Given that ALS-associated FUS mutants are deficient in nuclear localization, we examined whether cells expressing these mutants would be deficient in repressing exon 7 splicing. We showed that FUS harbouring R521G, R522G or ΔExon15 mutation (minor, moderate or severe cytoplasmic localization, respectively) directly correlated with respectively increasing deficiencies in both exon 7 repression and autoregulation of its own protein levels. These data suggest that compromised FUS autoregulation can directly exacerbate the pathogenic accumulation of cytoplasmic FUS protein in ALS. We showed that exon 7 skipping can be induced by antisense oligonucleotides targeting its flanking splice sites, indicating the potential to alleviate abnormal cytoplasmic FUS accumulation in ALS. Taken together, FUS autoregulation by alternative splicing provides insight into a molecular mechanism by which FUS-regulated pre-mRNA processing can impact a significant number of targets important to neurodegeneration.


Zdroje

1. AndersenPM, Al-ChalabiA (2011) Clinical genetics of amyotrophic lateral sclerosis: what do we really know? Nature Reviews Neurology 7: 603–615.

2. Da CruzS, ClevelandDW (2011) Understanding the role of TDP-43 and FUS/TLS in ALS and beyond. Current Opinion in Neurobiology 21: 904–919.

3. RobberechtW, PhilipsT (2013) The changing scene of amyotrophic lateral sclerosis. Nature Reviews Neuroscience 14: 248–264.

4. Lagier-TourenneC, ClevelandDW (2009) Rethinking ALS: The FUS about TDP-43. Cell 136: 1001–1004.

5. Lagier-TourenneC, PolymenidouM, ClevelandDW (2010) TDP-43 and FUS/TLS: emerging roles in RNA processing and neurodegeneration. Human Molecular Genetics 19: R46–R64.

6. KinoY, WashizuC, AquilantiE, OkunoM, KurosawaM, et al. (2010) Intracellular localization and splicing regulation of FUS/TLS are variably affected by amyotrophic lateral sclerosis-linked mutations. Nucleic Acids Research 39: 2781–2798.

7. DormannD, RoddeR, EdbauerD, BentmannE, FischerI, et al. (2010) ALS-associated fused in sarcoma (FUS) mutations disrupt Transportin-mediated nuclear import. The EMBO Journal 29: 2841–2857.

8. KwiatkowskiTJ, BoscoDA, LeClercAL, TamrazianE, VanderburgCR, et al. (2009) Mutations in the FUS/TLS Gene on Chromosome 16 Cause Familial Amyotrophic Lateral Sclerosis. Science 323: 1205–1208.

9. VanceC, RogeljB, HortobagyiT, De VosKJ, NishimuraAL, et al. (2009) Mutations in FUS, an RNA Processing Protein, Cause Familial Amyotrophic Lateral Sclerosis Type 6. Science 323: 1208–1211.

10. NeumannM, RademakersR, RoeberS, BakerM, KretzschmarHA, et al. (2009) A new subtype of frontotemporal lobar degeneration with FUS pathology. Brain 132: 2922–2931.

11. BoscoDA, LemayN, KoHK, ZhouH, BurkeC, et al. (2010) Mutant FUS proteins that cause amyotrophic lateral sclerosis incorporate into stress granules. Human Molecular Genetics 19: 4160–4175.

12. MackenzieIRA, AnsorgeO, StrongM, BilbaoJ, ZinmanL, et al. (2011) Pathological heterogeneity in amyotrophic lateral sclerosis with FUS mutations: two distinct patterns correlating with disease severity and mutation. Acta Neuropathologica 122: 87–98.

13. WaibelS, NeumannM, RosenbohmA, BirveA, VolkAE, et al. (2013) Truncating mutations in FUS/TLS give rise to a more aggressive ALS-phenotype than missense mutations: a clinico-genetic study in Germany. Eur J Neurol 20: 540–546.

14. FushimiK, LongC, JayaramN, ChenX, LiL, et al. (2011) Expression of human FUS/TLS in yeast leads to protein aggregation and cytotoxicity, recapitulating key features of FUS proteinopathy. Protein & Cell 2: 141–149.

15. ChenY, YangM, DengJ, ChenX, YeY, et al. (2011) Expression of human FUS protein in Drosophila leads to progressive neurodegeneration. Protein Cell 2: 477–486.

16. HuangC, ZhouH, TongJ, ChenH, LiuYJ, et al. (2011) FUS transgenic rats develop the phenotypes of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. PLoS Genet 7: e1002011.

17. MurakamiT, YangSP, XieL, KawanoT, FuD, et al. (2011) ALS mutations in FUS cause neuronal dysfunction and death in Caenorhabditis elegans by a dominant gain-of-function mechanism. Human Molecular Genetics 21: 1–9.

18. KabashiE, BercierV, LissoubaA, LiaoM, BrusteinE, et al. (2011) FUS and TARDBP but not SOD1 interact in genetic models of amyotrophic lateral sclerosis. PLoS Genet 7: e1002214.

19. IijimaKM, SasayamaH, ShimamuraM, TokudaT, AzumaY, et al. (2012) Knockdown of the Drosophila Fused in Sarcoma (FUS) Homologue Causes Deficient Locomotive Behavior and Shortening of Motoneuron Terminal Branches. PLoS One 7: e39483.

20. AnderssonMK, StåhlbergA, ArvidssonY, OlofssonA, SembH, et al. (2008) The multifunctional FUS, EWS and TAF15 proto-oncoproteins show cell type-specific expression patterns and involvement in cell spreading and stress response. BMC Cell Biology 9: 37.

21. ZinsznerH, SokJ, ImmanuelD, YinY, RonD (1997) TLS (FUS) binds RNA in vivo and engages in nucleo-cytoplasmic shuttling. J Cell Sci 110 Pt 15 1741–1750.

22. BaechtoldH, KurodaM, SokJ, RonD, LopezBS, et al. (1999) Human 75-kDa DNA-pairing protein is identical to the pro-oncoprotein TLS/FUS and is able to promote D-loop formation. J Biol Chem 274: 34337–34342.

23. LawWJ, CannKL, HicksGG (2006) TLS, EWS and TAF15: a model for transcriptional integration of gene expression. Brief Funct Genomic Proteomic 5: 8–14.

24. TanAY, ManleyJL (2009) The TET Family of Proteins: Functions and Roles in Disease. Journal of Molecular Cell Biology 1: 82–92.

25. DormannD, HaassC (2013) Fused in sarcoma (FUS): An oncogene goes awry in neurodegeneration. Mol Cell Neurosci E-pub ahead of print. doi: 10.1016/j.mcn.2013.03.006

26. AnthonyK, GalloJM (2010) Aberrant RNA processing events in neurological disorders. Brain Res 1338: 67–77.

27. RabinSJ, KimJM, BaughnM, LibbyRT, KimYJ, et al. (2010) Sporadic ALS has compartment-specific aberrant exon splicing and altered cell-matrix adhesion biology. Hum Mol Genet 19: 313–328.

28. IshigakiS, MasudaA, FujiokaY, IguchiY, KatsunoM, et al. (2012) Position-dependent FUS-RNA interactions regulate alternative splicing events and transcriptions. Sci Rep 2: 529.

29. RogeljB, EastonLE, BoguGK, StantonLW, RotG, et al. (2012) Widespread binding of FUS along nascent RNA regulates alternative splicing in the brain. Sci Rep 2: 603.

30. Lagier-TourenneC, PolymenidouM, HuttKR, VuAQ, BaughnM, et al. (2012) Divergent roles of ALS-linked proteins FUS/TLS and TDP-43 intersect in processing long pre-mRNAs. Nat Neurosci 15: 1488–1497.

31. HoellJI, LarssonE, RungeS, NusbaumJD, DuggimpudiS, et al. (2011) RNA targets of wild-type and mutant FET family proteins. Nature Structural & Molecular Biology 18: 1428–1431.

32. NakayaT, AlexiouP, MaragkakisM, ChangA, MourelatosZ (2013) FUS regulates genes coding for RNA-binding proteins in neurons by binding to their highly conserved introns. Rna 19: 498–509.

33. JiH, JiangH, MaW, JohnsonDS, MyersRM, et al. (2008) An integrated software system for analyzing ChIP-chip and ChIP-seq data. Nature Biotechnology 26: 1293–1300.

34. ColombritaC, OnestoE, MegiorniF, PizzutiA, BaralleFE, et al. (2012) TDP-43 and FUS RNA-binding Proteins Bind Distinct Sets of Cytoplasmic Messenger RNAs and Differently Regulate Their Post-transcriptional Fate in Motoneuron-like Cells. Journal of Biological Chemistry 287: 15635–15647.

35. XueY, ZhouY, WuT, ZhuT, JiX, et al. (2009) Genome-wide Analysis of PTB-RNA Interactions Reveals a Strategy Used by the General Splicing Repressor to Modulate Exon Inclusion or Skipping. Molecular Cell 36: 996–1006.

36. LicatalosiDD, MeleA, FakJJ, UleJ, KayikciM, et al. (2008) HITS-CLIP yields genome-wide insights into brain alternative RNA processing. Nature 456: 464–469.

37. HeinzS, BennerC, SpannN, BertolinoE, LinYC, et al. (2010) Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell 38: 576–589.

38. ZhangMQ (1998) Statistical features of human exons and their flanking regions. Hum Mol Genet 7: 919–932.

39. Huang daW, ShermanBT, LempickiRA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44–57.

40. LergaA, HallierM, DelvaL, OrvainC, GallaisI, et al. (2001) Identification of an RNA binding specificity for the potential splicing factor TLS. J Biol Chem 276: 6807–6816.

41. BoutzPL, StoilovP, LiQ, LinCH, ChawlaG, et al. (2007) A post-transcriptional regulatory switch in polypyrimidine tract-binding proteins reprograms alternative splicing in developing neurons. Genes & Development 21: 1636–1652.

42. XieJ, BlackDL (2001) A CaMK IV responsive RNA element mediates depolarization-induced alternative splicing of ion channels. Nature 410: 936–939.

43. RossbachO, HungLH, SchreinerS, GrishinaI, HeinerM, et al. (2009) Auto- and Cross-Regulation of the hnRNP L Proteins by Alternative Splicing. Molecular and Cellular Biology 29: 1442–1451.

44. MitchellJC, McGoldrickP, VanceC, HortobagyiT, SreedharanJ, et al. (2013) Overexpression of human wild-type FUS causes progressive motor neuron degeneration in an age- and dose-dependent fashion. Acta Neuropathol 125: 273–288.

45. SunZ, DiazZ, FangX, HartMP, ChesiA, et al. (2011) Molecular determinants and genetic modifiers of aggregation and toxicity for the ALS disease protein FUS/TLS. PLoS Biol 9: e1000614.

46. VanceC, ScotterEL, NishimuraAL, TroakesC, MitchellJC, et al. (2013) ALS mutant FUS disrupts nuclear localization and sequesters wild-type FUS within cytoplasmic stress granules. Human Molecular Genetics 22: 2676–2688.

47. SpitaliP, Aartsma-RusA (2012) Splice Modulating Therapies for Human Disease. Cell 148: 1085–1088.

48. WiltonSD, FallAM, HardingPL, McCloreyG, ColemanC, et al. (2007) Antisense Oligonucleotide-induced Exon Skipping Across the Human Dystrophin Gene Transcript. Molecular Therapy 15: 1288–1296.

49. HuaY, SahashiK, HungG, RigoF, PassiniMA, et al. (2010) Antisense correction of SMN2 splicing in the CNS rescues necrosis in a type III SMA mouse model. Genes & Development 24: 1634–1644.

50. WollertonMC, GoodingC, WagnerEJ, Garcia-BlancoMA, SmithCW (2004) Autoregulation of polypyrimidine tract binding protein by alternative splicing leading to nonsense-mediated decay. Mol Cell 13: 91–100.

51. DredgeBK, StefaniG, EngelhardCC, DarnellRB (2005) Nova autoregulation reveals dual functions in neuronal splicing. Embo J 24: 1608–1620.

52. AyalaYM, De ContiL, Avendaño-VázquezSE, DhirA, RomanoM, et al. (2010) TDP-43 regulates its mRNA levels through a negative feedback loop. The EMBO Journal 30: 277–288.

53. HicksGG, SinghN, NashabiA, MaiS, BozekG, et al. (2000) Fus deficiency in mice results in defective B-lymphocyte development and activation, high levels of chromosomal instability and perinatal death. Nat Genet 24: 175–179.

54. SpitzerJI, UgrasS, RungeS, DecarolisP, AntonescuC, et al. (2011) mRNA and protein levels of FUS, EWSR1, and TAF15 are upregulated in liposarcoma. Genes Chromosomes Cancer 50: 338–347.

55. MillsKI, WalshV, GilkesAF, SweeneyMC, MirzaT, et al. (2000) High FUS/TLS expression in acute myeloid leukaemia samples. Br J Haematol 108: 316–321.

56. PolymenidouM, Lagier-TourenneC, HuttKR, HuelgaSC, MoranJ, et al. (2011) Long pre-mRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43. Nature Neuroscience 14: 459–468.

57. LingS-C, PolymenidouM, Cleveland DonW (2013) Converging Mechanisms in ALS and FTD: Disrupted RNA and Protein Homeostasis. Neuron 79: 416–438.

58. SuzukiN, KatoS, KatoM, WaritaH, MizunoH, et al. (2012) FUS/TLS-immunoreactive neuronal and glial cell inclusions increase with disease duration in familial amyotrophic lateral sclerosis with an R521C FUS/TLS mutation. J Neuropathol Exp Neurol 71: 779–788.

59. KameokaS, DuqueP, KonarskaMM (2004) p54(nrb) associates with the 5′ splice site within large transcription/splicing complexes. Embo J 23: 1782–1791.

60. LeichterM, MarkoM, GanouV, Patrinou-GeorgoulaM, ToraL, et al. (2011) A fraction of the transcription factor TAF15 participates in interactions with a subset of the spliceosomal U1 snRNP complex. Biochim Biophys Acta 1814: 1812–1824.

61. YeoGW, CoufalNG, LiangTY, PengGE, FuXD, et al. (2009) An RNA code for the FOX2 splicing regulator revealed by mapping RNA-protein interactions in stem cells. Nat Struct Mol Biol 16: 130–137.

62. RayP, KarA, FushimiK, HavliogluN, ChenX, et al. (2011) PSF suppresses tau exon 10 inclusion by interacting with a stem-loop structure downstream of exon 10. J Mol Neurosci 45: 453–466.

63. HanK, YeoG, AnP, BurgeCB, GrabowskiPJ (2005) A combinatorial code for splicing silencing: UAGG and GGGG motifs. PLoS Biol 3: e158.

64. DichmannDS, HarlandRM (2012) fus/TLS orchestrates splicing of developmental regulators during gastrulation. Genes & Development 26: 1351–1363.

65. UleJ, JensenK, MeleA, DarnellRB (2005) CLIP: A method for identifying protein–RNA interaction sites in living cells. Methods 37: 376–386.

66. AltschulSF, GishW, MillerW, MyersEW, LipmanDJ (1990) Basic local alignment search tool. J Mol Biol 215: 403–410.

67. BlankenbergD, CoraorN, Von KusterG, TaylorJ, NekrutenkoA (2011) Integrating diverse databases into an unified analysis framework: a Galaxy approach. Database (Oxford) 2011: bar011.

68. LangmeadB, TrapnellC, PopM, SalzbergSL (2009) Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 10: R25.

69. FujitaPA, RheadB, ZweigAS, HinrichsAS, KarolchikD, et al. (2011) The UCSC Genome Browser database: update 2011. Nucleic Acids Res 39: D876–882.

70. SanfordJR, WangX, MortM, VanDuynN, CooperDN, et al. (2008) Splicing factor SFRS1 recognizes a functionally diverse landscape of RNA transcripts. Genome Research 19: 381–394.

71. NagarajN, WisniewskiJR, GeigerT, CoxJ, KircherM, et al. (2011) Deep proteome and transcriptome mapping of a human cancer cell line. Mol Syst Biol 7: 548.

72. ChenEY, TanCM, KouY, DuanQ, WangZ, et al. (2013) Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14: 128.

73. CooperC, GuoJ, YanY, Chooniedass-KothariS, HubeF, et al. (2009) Increasing the relative expression of endogenous non-coding Steroid Receptor RNA Activator (SRA) in human breast cancer cells using modified oligonucleotides. Nucleic Acids Research 37: 4518–4531.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 10
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#