#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Detection of Slipped-DNAs at the Trinucleotide Repeats of the Myotonic Dystrophy Type I Disease Locus in Patient Tissues


Slipped-strand DNAs, formed by out-of-register mispairing of repeat units on complementary strands, were proposed over 55 years ago as transient intermediates in repeat length mutations, hypothesized to cause at least 40 neurodegenerative diseases. While slipped-DNAs have been characterized in vitro, evidence of slipped-DNAs at an endogenous locus in biologically relevant tissues, where instability varies widely, is lacking. Here, using an anti-DNA junction antibody and immunoprecipitation, we identify slipped-DNAs at the unstable trinucleotide repeats (CTG)n•(CAG)n of the myotonic dystrophy disease locus in patient brain, heart, muscle and other tissues, where the largest expansions arise in non-mitotic tissues such as cortex and heart, and are smallest in the cerebellum. Slipped-DNAs are shown to be present on the expanded allele and in chromatinized DNA. Slipped-DNAs are present as clusters of slip-outs along a DNA, with each slip-out having 1–100 extrahelical repeats. The allelic levels of slipped-DNA containing molecules were significantly greater in the heart over the cerebellum (relative to genomic equivalents of pre-IP input DNA) of a DM1 individual; an enrichment consistent with increased allelic levels of slipped-DNA structures in tissues having greater levels of CTG instability. Surprisingly, this supports the formation of slipped-DNAs as persistent mutation products of repeat instability, and not merely as transient mutagenic intermediates. These findings further our understanding of the processes of mutation and genetic variation.


Vyšlo v časopise: Detection of Slipped-DNAs at the Trinucleotide Repeats of the Myotonic Dystrophy Type I Disease Locus in Patient Tissues. PLoS Genet 9(12): e32767. doi:10.1371/journal.pgen.1003866
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003866

Souhrn

Slipped-strand DNAs, formed by out-of-register mispairing of repeat units on complementary strands, were proposed over 55 years ago as transient intermediates in repeat length mutations, hypothesized to cause at least 40 neurodegenerative diseases. While slipped-DNAs have been characterized in vitro, evidence of slipped-DNAs at an endogenous locus in biologically relevant tissues, where instability varies widely, is lacking. Here, using an anti-DNA junction antibody and immunoprecipitation, we identify slipped-DNAs at the unstable trinucleotide repeats (CTG)n•(CAG)n of the myotonic dystrophy disease locus in patient brain, heart, muscle and other tissues, where the largest expansions arise in non-mitotic tissues such as cortex and heart, and are smallest in the cerebellum. Slipped-DNAs are shown to be present on the expanded allele and in chromatinized DNA. Slipped-DNAs are present as clusters of slip-outs along a DNA, with each slip-out having 1–100 extrahelical repeats. The allelic levels of slipped-DNA containing molecules were significantly greater in the heart over the cerebellum (relative to genomic equivalents of pre-IP input DNA) of a DM1 individual; an enrichment consistent with increased allelic levels of slipped-DNA structures in tissues having greater levels of CTG instability. Surprisingly, this supports the formation of slipped-DNAs as persistent mutation products of repeat instability, and not merely as transient mutagenic intermediates. These findings further our understanding of the processes of mutation and genetic variation.


Zdroje

1. Lopez CastelA, ClearyJD, PearsonCE (2010) Repeat instability as the basis for human diseases and as a potential target for therapy. Nat Rev Mol Cell Biol 11: 165–170.

2. PearsonCE, Nichol EdamuraK, ClearyJD (2005) Repeat instability: mechanisms of dynamic mutations. Nat Rev Genet 6: 729–742.

3. WangYH, AmirhaeriS, KangS, WellsRD, GriffithJD (1994) Preferential nucleosome assembly at DNA triplet repeats from the myotonic dystrophy gene. Science 265: 669–671.

4. Lopez CastelA, NakamoriM, TomeS, ChitayatD, GourdonG, et al. (2011) Expanded CTG repeat demarcates a boundary for abnormal CpG methylation in myotonic dystrophy patient tissues. Hum Mol Genet 20: 1–15.

5. ClearyJD, TomeS, Lopez CastelA, PanigrahiGB, FoiryL, et al. (2010) Tissue- and age-specific DNA replication patterns at the CTG/CAG-expanded human myotonic dystrophy type 1 locus. Nat Struct Mol Biol 17: 1079–1087.

6. PanigrahiGB, LauR, MontgomerySE, LeonardMR, PearsonCE (2005) Slipped (CTG)*(CAG) repeats can be correctly repaired, escape repair or undergo error-prone repair. Nat Struct Mol Biol 12: 654–662.

7. HouC, ChanNL, GuL, LiGM (2009) Incision-dependent and error-free repair of (CAG)(n)/(CTG)(n) hairpins in human cell extracts. Nat Struct Mol Biol 16: 869–875.

8. PearsonCE, SindenRR (1996) Alternative structures in duplex DNA formed within the trinucleotide repeats of the myotonic dystrophy and fragile X loci. Biochemistry 35: 5041–5053.

9. TamM, Erin MontgomeryS, KekisM, StollarBD, PriceGB, et al. (2003) Slipped (CTG).(CAG) repeats of the myotonic dystrophy locus: surface probing with anti-DNA antibodies. J Mol Biol 332: 585–600.

10. LiuG, ChenX, BisslerJJ, SindenRR, LeffakM (2010) Replication-dependent instability at (CTG) x (CAG) repeat hairpins in human cells. Nat Chem Biol 6: 652–659.

11. FrappierL, PriceGB, MartinRG, Zannis-HadjopoulosM (1989) Characterization of the binding specificity of two anticruciform DNA monoclonal antibodies. J Biol Chem 264: 334–341.

12. SteinmetzerK, Zannis-HadjopoulosM, PriceGB (1995) Anti-cruciform monoclonal antibody and cruciform DNA interaction. J Mol Biol 254: 29–37.

13. FelsenfeldG (1958) Theoretical Studies of the Interaction of synthetic polyribonucleotides. Biochim Biophys Acta 29: 133–144.

14. DeJesus-HernandezM, MackenzieIR, BoeveBF, BoxerAL, BakerM, et al. (2011) Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72: 245–256.

15. RentonAE, MajounieE, WaiteA, Simon-SanchezJ, RollinsonS, et al. (2011) A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72: 257–268.

16. PearsonCE, WangYH, GriffithJD, SindenRR (1998) Structural analysis of slipped-strand DNA (S-DNA) formed in (CTG)n. (CAG)n repeats from the myotonic dystrophy locus. Nucleic Acids Res 26: 816–823.

17. PearsonCE, TamM, WangYH, MontgomerySE, DarAC, et al. (2002) Slipped-strand DNAs formed by long (CAG)*(CTG) repeats: slipped-out repeats and slip-out junctions. Nucleic Acids Res 30: 4534–4547.

18. SleanMM, ReddyK, WuB, Nichol EdamuraK, KekisM, et al. (2013) Interconverting conformations of slipped-DNA junctions formed by trinucleotide repeats affect repair outcome. Biochemistry 52: 773–785.

19. FrappierL, PriceGB, MartinRG, Zannis-HadjopoulosM (1987) Monoclonal antibodies to cruciform DNA structures. J Mol Biol 193: 751–758.

20. BacollaA, GellibolianR, ShimizuM, AmirhaeriS, KangS, et al. (1997) Flexible DNA: genetically unstable CTG.CAG and CGG.CCG from human hereditary neuromuscular disease genes. J Biol Chem 272: 16783–16792.

21. WarnerJP, BarronLH, GoudieD, KellyK, DowD, et al. (1996) A general method for the detection of large CAG repeat expansions by fluorescent PCR. J Med Genet 33: 1022–1026.

22. ZentilinL, GiaccaM (2007) Competitive PCR for precise nucleic acid quantification. Nat Protoc 2: 2092–2104.

23. ZentilinL, GiaccaM (2010) The renaissance of competitive PCR as an accurate tool for precise nucleic acid quantification. Methods Mol Biol 630: 233–248.

24. GaubatzJ, EllisM, ChalkleyR (1979) Nuclease digestion studies of mouse chromatin as a function of age. J Gerontol 34: 672–679.

25. LarsenA, WeintraubH (1982) An altered DNA conformation detected by S1 nuclease occurs at specific regions in active chick globin chromatin. Cell 29: 609–622.

26. NickolJ, MartinRG (1983) DNA stem-loop structures bind poorly to histone octamer cores. Proc Natl Acad Sci U S A 80: 4669–4673.

27. NobileC, NickolJ, MartinRG (1986) Nucleosome phasing on a DNA fragment from the replication origin of simian virus 40 and rephasing upon cruciform formation of the DNA. Mol Cell Biol 6: 2916–2922.

28. PanigrahiGB, SleanMM, SimardJP, GileadiO, PearsonCE (2010) Isolated short CTG/CAG DNA slip-outs are repaired efficiently by hMutSbeta, but clustered slip-outs are poorly repaired. Proc Natl Acad Sci U S A 107: 12593–12598.

29. FreudenreichCH, KantrowSM, ZakianVA (1998) Expansion and length-dependent fragility of CTG repeats in yeast. Science 279: 853–856.

30. MarcadierJL, PearsonCE (2003) Fidelity of primate cell repair of a double-strand break within a (CTG).(CAG) tract. Effect of slipped DNA structures. J Biol Chem 278: 33848–33856.

31. DobbingJ, SandsJ (1979) Comparative aspects of the brain growth spurt. Early Hum Dev 3: 79–83.

32. ten DonkelaarHJ, LammensM, WesselingP, ThijssenHO, RenierWO (2003) Development and developmental disorders of the human cerebellum. J Neurol 250: 1025–1036.

33. ShankleWR, LandingBH, RafiiMS, SchianoA, ChenJM, et al. (1998) Evidence for a postnatal doubling of neuron number in the developing human cerebral cortex between 15 months and 6 years. J Theor Biol 191: 115–140.

34. Cruz-MunozW, KalairW, CosentinoL, HeddleJA (2000) ENU induces mutations in the heart of lacZ transgenic mice. Mutat Res 469: 23–34.

35. EngermanRL, PfaffenbachD, DavisMD (1967) Cell turnover of capillaries. Lab Invest 17: 738–743.

36. MarinoTA, HaldarS, WilliamsonEC, BeaversonK, WalterRA, et al. (1991) Proliferating cell nuclear antigen in developing and adult rat cardiac muscle cells. Circ Res 69: 1353–1360.

37. TeleniusH, KremerB, GoldbergYP, TheilmannJ, AndrewSE, et al. (1994) Somatic and gonadal mosaicism of the Huntington disease gene CAG repeat in brain and sperm. Nat Genet 6: 409–414.

38. De RooijKE, De Koning GansPA, RoosRA, Van OmmenGJ, Den DunnenJT (1995) Somatic expansion of the (CAG)n repeat in Huntington disease brains. Hum Genet 95: 270–274.

39. OnoderaO, IdezukaJ, IgarashiS, TakiyamaY, EndoK, et al. (1998) Progressive atrophy of cerebellum and brainstem as a function of age and the size of the expanded CAG repeats in the MJD1 gene in Machado-Joseph disease. Ann Neurol 43: 288–296.

40. RomeoV, PegoraroE, FerratiC, SquarzantiF, SoraruG, et al. (2010) Brain involvement in myotonic dystrophies: neuroimaging and neuropsychological comparative study in DM1 and DM2. J Neurol 257: 1246–1255.

41. LeroyO, WangJ, MaurageCA, ParentM, CooperT, et al. (2006) Brain-specific change in alternative splicing of Tau exon 6 in myotonic dystrophy type 1. Biochim Biophys Acta 1762: 460–467.

42. GrohWJ, LoweMR, ZipesDP (2002) Severity of cardiac conduction involvement and arrhythmias in myotonic dystrophy type 1 correlates with age and CTG repeat length. J Cardiovasc Electrophysiol 13: 444–448.

43. ThorntonCA, JohnsonK, MoxleyRT3rd (1994) Myotonic dystrophy patients have larger CTG expansions in skeletal muscle than in leukocytes. Ann Neurol 35: 104–107.

44. StreisingerG, OkadaY, EmrichJ, NewtonJ, TsugitaA, et al. (1966) Frameshift mutations and the genetic code. This paper is dedicated to Professor Theodosius Dobzhansky on the occasion of his 66th birthday. Cold Spring Harb Symp Quant Biol 31: 77–84.

45. BergI, NeumannR, CederbergH, RannugU, JeffreysAJ (2003) Two modes of germline instability at human minisatellite MS1 (locus D1S7): complex rearrangements and paradoxical hyperdeletion. Am J Hum Genet 72: 1436–1447.

46. BoyerJC, BebenekK, KunkelTA (1992) Unequal human immunodeficiency virus type 1 reverse transcriptase error rates with RNA and DNA templates. Proc Natl Acad Sci U S A 89: 6919–6923.

47. BuardJ, CollickA, BrownJ, JeffreysAJ (2000) Somatic versus germline mutation processes at minisatellite CEB1 (D2S90) in humans and transgenic mice. Genomics 65: 95–103.

48. GugliettaS, PantaleoG, GraziosiC (2010) Long sequence duplications, repeats, and palindromes in HIV-1 gp120: length variation in V4 as the product of misalignment mechanism. Virology 399: 167–175.

49. WangL, ParadeeW, MullinsC, ShridharR, RosatiR, et al. (1997) Aphidicolin-induced FRA3B breakpoints cluster in two distinct regions. Genomics 41: 485–488.

50. DiviaccoS, NorioP, ZentilinL, MenzoS, ClementiM, et al. (1992) A novel procedure for quantitative polymerase chain reaction by coamplification of competitive templates. Gene 122: 313–320.

51. MenzoS, BagnarelliP, GiaccaM, ManzinA, VaraldoPE, et al. (1992) Absolute quantitation of viremia in human immunodeficiency virus infection by competitive reverse transcription and polymerase chain reaction. J Clin Microbiol 30: 1752–1757.

52. SestiniR, OrlandoC, ZentilinL, GelminiS, PinzaniP, et al. (1994) Measuring c-erbB-2 oncogene amplification in fresh and paraffin-embedded tumors by competitive polymerase chain reaction. Clin Chem 40: 630–636.

53. GiaccaM, ZentilinL, NorioP, DiviaccoS, DimitrovaD, et al. (1994) Fine mapping of a replication origin of human DNA. Proc Natl Acad Sci U S A 91: 7119–7123.

54. PelizonC, DiviaccoS, FalaschiA, GiaccaM (1996) High-resolution mapping of the origin of DNA replication in the hamster dihydrofolate reductase gene domain by competitive PCR. Mol Cell Biol 16: 5358–5364.

55. AbdurashidovaG, DanailovMB, OchemA, TrioloG, DjeliovaV, et al. (2003) Localization of proteins bound to a replication origin of human DNA along the cell cycle. EMBO J 22: 4294–4303.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 12
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#