#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

MSH3 Polymorphisms and Protein Levels Affect CAG Repeat Instability in Huntington's Disease Mice


Expansions of trinucleotide CAG/CTG repeats in somatic tissues are thought to contribute to ongoing disease progression through an affected individual's life with Huntington's disease or myotonic dystrophy. Broad ranges of repeat instability arise between individuals with expanded repeats, suggesting the existence of modifiers of repeat instability. Mice with expanded CAG/CTG repeats show variable levels of instability depending upon mouse strain. However, to date the genetic modifiers underlying these differences have not been identified. We show that in liver and striatum the R6/1 Huntington's disease (HD) (CAG)∼100 transgene, when present in a congenic C57BL/6J (B6) background, incurred expansion-biased repeat mutations, whereas the repeat was stable in a congenic BALB/cByJ (CBy) background. Reciprocal congenic mice revealed the Msh3 gene as the determinant for the differences in repeat instability. Expansion bias was observed in congenic mice homozygous for the B6 Msh3 gene on a CBy background, while the CAG tract was stabilized in congenics homozygous for the CBy Msh3 gene on a B6 background. The CAG stabilization was as dramatic as genetic deficiency of Msh2. The B6 and CBy Msh3 genes had identical promoters but differed in coding regions and showed strikingly different protein levels. B6 MSH3 variant protein is highly expressed and associated with CAG expansions, while the CBy MSH3 variant protein is expressed at barely detectable levels, associating with CAG stability. The DHFR protein, which is divergently transcribed from a promoter shared by the Msh3 gene, did not show varied levels between mouse strains. Thus, naturally occurring MSH3 protein polymorphisms are modifiers of CAG repeat instability, likely through variable MSH3 protein stability. Since evidence supports that somatic CAG instability is a modifier and predictor of disease, our data are consistent with the hypothesis that variable levels of CAG instability associated with polymorphisms of DNA repair genes may have prognostic implications for various repeat-associated diseases.


Vyšlo v časopise: MSH3 Polymorphisms and Protein Levels Affect CAG Repeat Instability in Huntington's Disease Mice. PLoS Genet 9(2): e32767. doi:10.1371/journal.pgen.1003280
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003280

Souhrn

Expansions of trinucleotide CAG/CTG repeats in somatic tissues are thought to contribute to ongoing disease progression through an affected individual's life with Huntington's disease or myotonic dystrophy. Broad ranges of repeat instability arise between individuals with expanded repeats, suggesting the existence of modifiers of repeat instability. Mice with expanded CAG/CTG repeats show variable levels of instability depending upon mouse strain. However, to date the genetic modifiers underlying these differences have not been identified. We show that in liver and striatum the R6/1 Huntington's disease (HD) (CAG)∼100 transgene, when present in a congenic C57BL/6J (B6) background, incurred expansion-biased repeat mutations, whereas the repeat was stable in a congenic BALB/cByJ (CBy) background. Reciprocal congenic mice revealed the Msh3 gene as the determinant for the differences in repeat instability. Expansion bias was observed in congenic mice homozygous for the B6 Msh3 gene on a CBy background, while the CAG tract was stabilized in congenics homozygous for the CBy Msh3 gene on a B6 background. The CAG stabilization was as dramatic as genetic deficiency of Msh2. The B6 and CBy Msh3 genes had identical promoters but differed in coding regions and showed strikingly different protein levels. B6 MSH3 variant protein is highly expressed and associated with CAG expansions, while the CBy MSH3 variant protein is expressed at barely detectable levels, associating with CAG stability. The DHFR protein, which is divergently transcribed from a promoter shared by the Msh3 gene, did not show varied levels between mouse strains. Thus, naturally occurring MSH3 protein polymorphisms are modifiers of CAG repeat instability, likely through variable MSH3 protein stability. Since evidence supports that somatic CAG instability is a modifier and predictor of disease, our data are consistent with the hypothesis that variable levels of CAG instability associated with polymorphisms of DNA repair genes may have prognostic implications for various repeat-associated diseases.


Zdroje

1. LeeJM, RamosEM, LeeJH, GillisT, MysoreJS, et al. (2012) CAG repeat expansion in Huntington disease determines age at onset in a fully dominant fashion. Neurology 78: 690–695.

2. RosenblattA, KumarBV, MoA, WelshCS, MargolisRL, et al. (2012) Age, CAG repeat length, and clinical progression in Huntington's disease. Mov Disord 27: 272–276.

3. 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.

4. 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.

5. KennedyL, EvansE, ChenCM, CravenL, DetloffPJ, et al. (2003) Dramatic tissue-specific mutation length increases are an early molecular event in Huntington disease pathogenesis. Hum Mol Genet 12: 3359–3367.

6. ShelbournePF, Keller-McGandyC, BiWL, YoonSR, DubeauL, et al. (2007) Triplet repeat mutation length gains correlate with cell-type specific vulnerability in Huntington disease brain. Hum Mol Genet 16: 1133–1142.

7. SwamiM, HendricksAE, GillisT, MassoodT, MysoreJ, et al. (2009) Somatic expansion of the Huntington's disease CAG repeat in the brain is associated with an earlier age of disease onset. Hum Mol Genet 18: 3039–3047.

8. MoralesF, CoutoJM, HighamCF, HoggG, CuencaP, et al. (2012) Somatic instability of the expanded CTG triplet repeat in myotonic dystrophy type 1 is a heritable quantitative trait and modifier of disease severity. Hum Mol Genet 21: 3558–3567.

9. 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.

10. MangiariniL, SathasivamK, MahalA, MottR, SellerM, et al. (1997) Instability of highly expanded CAG repeats in mice transgenic for the Huntington's disease mutation. Nat Genet 15: 197–200.

11. GourdonG, RadvanyiF, LiaAS, DurosC, BlancheM, et al. (1997) Moderate intergenerational and somatic instability of a 55-CTG repeat in transgenic mice. Nat Genet 15: 190–192.

12. MoncktonDG, CoolbaughMI, AshizawaKT, SicilianoMJ, CaskeyCT (1997) Hypermutable myotonic dystrophy CTG repeats in transgenic mice. Nat Genet 15: 193–196.

13. ManleyK, ShirleyTL, FlahertyL, MesserA (1999) Msh2 deficiency prevents in vivo somatic instability of the CAG repeat in Huntington disease transgenic mice. Nat Genet 23: 471–473.

14. ShelbournePF, KilleenN, HevnerRF, JohnstonHM, TecottL, et al. (1999) A Huntington's disease CAG expansion at the murine Hdh locus is unstable and associated with behavioural abnormalities in mice. Hum Mol Genet 8: 763–774.

15. WheelerVC, AuerbachW, WhiteJK, SrinidhiJ, AuerbachA, et al. (1999) Length-dependent gametic CAG repeat instability in the Huntington's disease knock-in mouse. Hum Mol Genet 8: 115–122.

16. van Den BroekWJ, NelenMR, WansinkDG, CoerwinkelMM, te RieleH, et al. (2002) Somatic expansion behaviour of the (CTG)(n) repeat in myotonic dystrophy knock-in mice is differentially affected by Msh3 and Msh6 mismatch-repair proteins. Hum Mol Genet 11: 191–198.

17. LibbyRT, MoncktonDG, FuYH, MartinezRA, McAbneyJP, et al. (2003) Genomic context drives SCA7 CAG repeat instability, while expressed SCA7 cDNAs are intergenerationally and somatically stable in transgenic mice. Hum Mol Genet 12: 41–50.

18. LibbyRT, HagermanKA, PinedaVV, LauR, ChoDH, et al. (2008) CTCF cis-regulates trinucleotide repeat instability in an epigenetic manner: a novel basis for mutational hot spot determination. PLoS Genet 4: e1000257 doi:10.1371/journal.pgen.1000257.

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

20. WarbySC, MontpetitA, HaydenAR, CarrollJB, ButlandSL, et al. (2009) CAG expansion in the Huntington disease gene is associated with a specific and targetable predisposing haplogroup. Am J Hum Genet 84: 351–366.

21. BrockGJ, AndersonNH, MoncktonDG (1999) Cis-acting modifiers of expanded CAG/CTG triplet repeat expandability: associations with flanking GC content and proximity to CpG islands. Hum Mol Genet 8: 1061–1067.

22. NestorCE, MoncktonDG (2011) Correlation of inter-locus polyglutamine toxicity with CAG•CTG triplet repeat expandability and flanking genomic DNA GC content. PLoS ONE 6: e28260 doi:10.1371/journal.pone.0028260.

23. ClearyJD, PearsonCE (2003) The contribution of cis-elements to disease-associated repeat instability: Clinical and experimental evidence. Cytogenetics and Genome Research 100: 25–55.

24. van den BroekWJ, NelenMR, van der HeijdenGW, WansinkDG, WieringaB (2006) Fen1 does not control somatic hypermutability of the (CTG)(n)•(CAG)(n) repeat in a knock-in mouse model for DM1. FEBS Lett 580: 5208–5214.

25. MollersenL, RoweAD, LarsenE, RognesT, KlunglandA (2010) Continuous and periodic expansion of CAG repeats in Huntington's disease R6/1 mice. PLoS Genet 6: e1001242 doi:10.1371/journal.pgen.1001242.

26. SavouretC, BrissonE, EssersJ, KanaarR, PastinkA, et al. (2003) CTG repeat instability and size variation timing in DNA repair-deficient mice. Embo J 22: 2264–2273.

27. DragilevaE, HendricksA, TeedA, GillisT, LopezET, et al. (2009) Intergenerational and striatal CAG repeat instability in Huntington's disease knock-in mice involve different DNA repair genes. Neurobiol Dis 33: 37–47.

28. Gomes-PereiraM, FortuneMT, IngramL, McAbneyJP, MoncktonDG (2004) Pms2 is a genetic enhancer of trinucleotide CAG.CTG repeat somatic mosaicism: implications for the mechanism of triplet repeat expansion. Hum Mol Genet 13: 1815–1825.

29. TomeS, PanigrahiGB, Lopez CastelA, FoiryL, MeltonDW, et al. (2011) Maternal germline-specific effect of DNA ligase I on CTG/CAG instability. Hum Mol Genet 20: 2131–2143.

30. HubertLJr, LinY, DionV, WilsonJH (2011) Xpa deficiency reduces CAG trinucleotide repeat instability in neuronal tissues in a mouse model of SCA1. Hum Mol Genet 20: 4822–4830.

31. MollersenL, RoweAD, IlluzziJL, HildrestrandGA, GerholdKJ, et al. (2012) Neil1 is a genetic modifier of somatic and germline CAG trinucleotide repeat instability in R6/1 mice. Hum Mol Genet

32. KovtunIV, JohnsonKO, McMurrayCT (2011) Cockayne syndrome B protein antagonizes OGG1 in modulating CAG repeat length in vivo. Aging (Albany NY) 3: 509–514.

33. KovtunIV, LiuY, BjorasM, KlunglandA, WilsonSH, et al. (2007) OGG1 initiates age-dependent CAG trinucleotide expansion in somatic cells. Nature 447: 447–452.

34. SavouretC, Garcia-CordierC, MegretJ, te RieleH, JunienC, et al. (2004) MSH2-dependent germinal CTG repeat expansions are produced continuously in spermatogonia from DM1 transgenic mice. Mol Cell Biol 24: 629–637.

35. FoiryL, DongL, SavouretC, HubertL, te RieleH, et al. (2006) Msh3 is a limiting factor in the formation of intergenerational CTG expansions in DM1 transgenic mice. Hum Genet 119: 520–526.

36. WheelerVC, LebelLA, VrbanacV, TeedA, Te RieleH, et al. (2003) Mismatch repair gene Msh2 modifies the timing of early disease in Hdh(Q111) striatum. Hum Mol Genet 12: 273–281.

37. SleanMM, PanigrahiGB, RanumLP, PearsonCE (2008) Mutagenic roles of DNA “repair” proteins in antibody diversity and disease-associated trinucleotide repeat instability. DNA Repair (Amst) 7: 1135–1154.

38. SeriolaA, SpitsC, SimardJP, HilvenP, HaentjensP, et al. (2011) Huntington's and myotonic dystrophy hESCs: down-regulated trinucleotide repeat instability and mismatch repair machinery expression upon differentiation. Hum Mol Genet 20: 176–185.

39. JiricnyJ (2006) The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol 7: 335–346.

40. GenschelJ, LittmanSJ, DrummondJT, ModrichP (1998) Isolation of MutSbeta from human cells and comparison of the mismatch repair specificities of MutSbeta and MutSalpha. J Biol Chem 273: 19895–19901.

41. LittmanSJ, FangWH, ModrichP (1999) Repair of large insertion/deletion heterologies in human nuclear extracts is directed by a 5′ single-strand break and is independent of the mismatch repair system. J Biol Chem 274: 7474–7481.

42. 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.

43. HarringtonJM, KolodnerRD (2007) Saccharomyces cerevisiae Msh2-Msh3 acts in repair of base-base mispairs. Mol Cell Biol 27: 6546–6554.

44. TianL, GuL, LiGM (2009) Distinct nucleotide binding/hydrolysis properties and molar ratio of MutSalpha and MutSbeta determine their differential mismatch binding activities. J Biol Chem 284: 11557–11562.

45. TomeS, SimardJP, SleanMM, HoltI, MorrisGE, et al. (2013) Tissue-specific mismatch repair protein expression: MSH3 is higher than MSH6 in multiple mouse tissues. DNA Repair (Amst) 12: 46–52.

46. ManleyK, PughJ, MesserA (1999) Instability of the CAG repeat in immortalized fibroblast cell cultures from Huntington's disease transgenic mice. Brain Res 835: 74–79.

47. TomeS, HoltI, EdelmannW, MorrisGE, MunnichA, et al. (2009) MSH2 ATPase domain mutation affects CTG•CAG repeat instability in transgenic mice. PLoS Genet 5: e1000482 doi:10.1371/journal.pgen.100048.

48. ChangDK, RicciardielloL, GoelA, ChangCL, BolandCR (2000) Steady-state regulation of the human DNA mismatch repair system. J Biol Chem 275: 29178.

49. KovtunIV, ThornhillAR, McMurrayCT (2004) Somatic deletion events occur during early embryonic development and modify the extent of CAG expansion in subsequent generations. Hum Mol Genet 13: 3057–3068.

50. PearsonCE, EwelA, AcharyaS, FishelRA, SindenRR (1997) Human MSH2 binds to trinucleotide repeat DNA structures associated with neurodegenerative diseases. Hum Mol Genet 6: 1117–1123.

51. 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.

52. LinY, DionV, WilsonJH (2006) Transcription promotes contraction of CAG repeat tracts in human cells. Nat Struct Mol Biol 13: 179–180.

53. Lopez CastelA, TomkinsonAE, PearsonCE (2009) CTG/CAG repeat instability is modulated by the levels of human DNA ligase i and its interaction with proliferating cell nuclear antigen: a distinction between replication and slipped-DNA repair. J Biol Chem 284: 26631–26645.

54. GoulaAV, BerquistBR, WilsonDM3rd, WheelerVC, TrottierY, et al. (2009) Stoichiometry of base excision repair proteins correlates with increased somatic CAG instability in striatum over cerebellum In Huntington's disease transgenic mice. PLoS Genet 5: e1000749 doi:10.1371/journal.pgen.1000749.

55. GoulaAV, PearsonCE, Della MariaJ, TrottierY, TomkinsonAE, et al. (2012) The nucleotide sequence, DNA damage location, and protein stoichiometry influence the base excision repair outcome at CAG/CTG repeats. Biochemistry 51: 3919–3932.

56. MangiariniL, SathasivamK, SellerM, CozensB, HarperA, et al. (1996) Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 87: 493–506.

57. ChiangC, JacobsenJC, ErnstC, HanscomC, HeilbutA, et al. (2012) Complex reorganization and predominant non-homologous repair following chromosomal breakage in karyotypically balanced germline rearrangements and transgenic integration. Nat Genet 44: 390–397, S391.

58. GoulaAV, StysA, ChanJP, TrottierY, FestensteinR, et al. (2012) Transcription elongation and tissue-specific somatic CAG instability. PLoS Genet 8: e1003051 doi:10.1371/journal.pgen.1003051.

59. LloretA, DragilevaE, TeedA, EspinolaJ, FossaleE, et al. (2006) Genetic background modifies nuclear mutant huntingtin accumulation and HD CAG repeat instability in Huntington's disease knock-in mice. Hum Mol Genet 15: 2015–2024.

60. CowinRM, BuiN, GrahamD, GreenJR, GrueningerS, et al. (2011) Onset and progression of behavioral and molecular phenotypes in a novel congenic R6/2 line exhibiting intergenerational CAG repeat stability. PLoS ONE 6: e28409 doi:10.1371/journal.pone.0028409.

61. RamosEM, CerqueiraJ, LemosC, Pinto-BastoJ, AlonsoI, et al. (2012) Intergenerational instability in Huntington disease: extreme repeat changes among 134 transmissions. Mov Disord 27: 583–585.

62. ZhangY, MoncktonDG, SicilianoMJ, ConnorTH, MeistrichML (2002) Age and insertion site dependence of repeat number instability of a human DM1 transgene in individual mouse sperm. Hum Mol Genet 11: 791–798.

63. HaugeXY, LittM (1993) A study of the origin of ‘shadow bands’ seen when typing dinucleotide repeat polymorphisms by the PCR. Hum Mol Genet 2: 411–415.

64. VatsavayaiSC, DalleracGM, MilnerwoodAJ, CummingsDM, RezaieP, et al. (2007) Progressive CAG expansion in the brain of a novel R6/1-89Q mouse model of Huntington's disease with delayed phenotypic onset. Brain Res Bull 72: 98–102.

65. HoltI, Thanh LamL, TomeS, WansinkDG, Te RieleH, et al. (2011) The mouse mismatch repair protein, MSH3, is a nucleoplasmic protein that aggregates into denser nuclear bodies under conditions of stress. J Cell Biochem 112: 1612–1621.

66. CrouseGF, LeysEJ, McEwanRN, FrayneEG, KellemsRE (1985) Analysis of the mouse dhfr promoter region: existence of a divergently transcribed gene. Mol Cell Biol 5: 1847–1858.

67. LintonJP, YenJY, SelbyE, ChenZ, ChinskyJM, et al. (1989) Dual bidirectional promoters at the mouse dhfr locus: cloning and characterization of two mRNA classes of the divergently transcribed Rep-1 gene. Mol Cell Biol 9: 3058–3072.

68. WatanabeA, IkejimaM, SuzukiN, ShimadaT (1996) Genomic organization and expression of the human MSH3 gene. Genomics 31: 311–318.

69. HutchinsonEG, ThorntonJM (1994) A revised set of potentials for beta-turn formation in proteins. Protein Sci 3: 2207–2216.

70. LamersMH, PerrakisA, EnzlinJH, WinterwerpHH, de WindN, et al. (2000) The crystal structure of DNA mismatch repair protein MutS binding to a G x T mismatch. Nature 407: 711–717.

71. TakanoK, YamagataY, YutaniK (2000) Role of amino acid residues at turns in the conformational stability and folding of human lysozyme. Biochemistry 39: 8655–8665.

72. MarcelinoAM, GieraschLM (2008) Roles of beta-turns in protein folding: from peptide models to protein engineering. Biopolymers 89: 380–391.

73. FullerAA, DuD, LiuF, DavorenJE, BhabhaG, et al. (2009) Evaluating beta-turn mimics as beta-sheet folding nucleators. Proc Natl Acad Sci U S A 106: 11067–11072.

74. SavouretC, JunienC, GourdonG (2004) Analysis of CTG Repeats Using DM1 Model Mice. Methods Mol Biol 277: 185–198.

75. FuH, GrimsleyGR, RazviA, ScholtzJM, PaceCN (2009) Increasing protein stability by improving beta-turns. Proteins 77: 491–498.

76. TrevinoSR, ScholtzJM, PaceCN (2007) Amino acid contribution to protein solubility: Asp, Glu, and Ser contribute more favorably than the other hydrophilic amino acids in RNase Sa. J Mol Biol 366: 449–460.

77. ChenRP, HuangJJ, ChenHL, JanH, VelusamyM, et al. (2004) Measuring the refolding of beta-sheets with different turn sequences on a nanosecond time scale. Proc Natl Acad Sci U S A 101: 7305–7310.

78. McCallisterEL, AlmE, BakerD (2000) Critical role of beta-hairpin formation in protein G folding. Nat Struct Biol 7: 669–673.

79. YbeJA, HechtMH (1996) Sequence replacements in the central beta-turn of plastocyanin. Protein Sci 5: 814–824.

80. OlsenJV, VermeulenM, SantamariaA, KumarC, MillerML, et al. (2010) Quantitative phosphoproteomics reveals widespread full phosphorylation site occupancy during mitosis. Sci Signal 3: ra3.

81. JohnsonLN, LewisRJ (2001) Structural basis for control by phosphorylation. Chem Rev 101: 2209–2242.

82. TakanoH, OnoderaO, TakahashiH, IgarashiS, YamadaM, et al. (1996) Somatic mosaicism of expanded CAG repeats in brains of patients with dentatorubral-pallidoluysian atrophy: cellular population-dependent dynamics of mitotic instability. Am J Hum Genet 58: 1212–1222.

83. WatanabeY, Haugen-StranoA, UmarA, YamadaK, HemmiH, et al. (2000) Complementation of an hMSH2 defect in human colorectal carcinoma cells by human chromosome 2 transfer. Mol Carcinog 29: 37–49.

84. HashidaH, GotoJ, SuzukiT, JeongS, MasudaN, et al. (2001) Single cell analysis of CAG repeat in brains of dentatorubral-pallidoluysian atrophy (DRPLA). J Neurol Sci 190: 87–93.

85. OdaS, MaeharaY, IkedaY, OkiE, EgashiraA, et al. (2005) Two modes of microsatellite instability in human cancer: differential connection of defective DNA mismatch repair to dinucleotide repeat instability. Nucleic Acids Res 33: 1628–1636.

86. GiuntiL, CeticaV, RicciU, GiglioS, SardiI, et al. (2009) Type A microsatellite instability in pediatric gliomas as an indicator of Turcot syndrome. Eur J Hum Genet 17: 919–927.

87. ThibodeauSN, BrenG, SchaidD (1993) Microsatellite instability in cancer of the proximal colon. Science 260: 816–819.

88. YangZ, LauR, MarcadierJL, ChitayatD, PearsonCE (2003) Replication inhibitors modulate instability of an expanded trinucleotide repeat at the myotonic dystrophy type 1 disease locus in human cells. Am J Hum Genet 73: 1092–1105.

89. WeberJL (1990) Informativeness of human (dC-dA)n.(dG-dT)n polymorphisms. Genomics 7: 524–530.

90. BlakeC, TsaoJL, WuA, ShibataD (2001) Stepwise deletions of polyA sequences in mismatch repair-deficient colorectal cancers. Am J Pathol 158: 1867–1870.

91. OttoCJ, AlmqvistE, HaydenMR, AndrewSE (2001) The “flap” endonuclease gene FEN1 is excluded as a candidate gene implicated in the CAG repeat expansion underlying Huntington disease. Clin Genet 59: 122–127.

92. CoppedeF, MigheliF, CeravoloR, BregantE, RocchiA, et al. (2010) The hOGG1 Ser326Cys polymorphism and Huntington's disease. Toxicology 278: 199–203.

93. Taherzadeh-FardE, SaftC, WieczorekS, EpplenJT, ArningL (2010) Age at onset in Huntington's disease: replication study on the associations of ADORA2A, HAP1 and OGG1. Neurogenetics 11: 435–439.

94. PanigrahiGB, SleanMM, SimardJP, PearsonCE (2012) Human Mismatch Repair Protein hMutLalpha Is Required to Repair Short Slipped-DNAs of Trinucleotide Repeats. J Biol Chem 287: 41844–41850.

95. MartinezSL, KolodnerRD (2010) Functional analysis of human mismatch repair gene mutations identifies weak alleles and polymorphisms capable of polygenic interactions. Proc Natl Acad Sci U S A 107: 5070–5075.

96. KumarC, PiacenteSC, SibertJ, BukataAR, O'ConnorJ, et al. (2011) Multiple factors insulate Msh2–Msh6 mismatch repair activity from defects in Msh2 domain I. J Mol Biol 411: 765–780.

97. MangoniM, BisanziS, CarozziF, SaniC, BitiG, et al. (2011) Association between genetic polymorphisms in the XRCC1, XRCC3, XPD, GSTM1, GSTT1, MSH2, MLH1, MSH3, and MGMT genes and radiosensitivity in breast cancer patients. Int J Radiat Oncol Biol Phys 81: 52–58.

98. DongX, LiY, HessKR, AbbruzzeseJL, LiD (2011) DNA mismatch repair gene polymorphisms affect survival in pancreatic cancer. Oncologist 16: 61–70.

99. CondeJ, SilvaSN, AzevedoAP, TeixeiraV, PinaJE, et al. (2009) Association of common variants in mismatch repair genes and breast cancer susceptibility: a multigene study. BMC Cancer 9: 344.

100. MichielsS, DanoyP, DessenP, BeraA, BouletT, et al. (2007) Polymorphism discovery in 62 DNA repair genes and haplotype associations with risks for lung and head and neck cancers. Carcinogenesis 28: 1731–1739.

101. HaugenAC, GoelA, YamadaK, MarraG, NguyenTP, et al. (2008) Genetic instability caused by loss of MutS homologue 3 in human colorectal cancer. Cancer Res 68: 8465–8472.

102. BurrKL, van Duyn-GoedhartA, HickenbothamP, MongerK, van BuulPP, et al. (2007) The effects of MSH2 deficiency on spontaneous and radiation-induced mutation rates in the mouse germline. Mutat Res 617: 147–151.

103. DuJ, CampauE, SoragniE, KuS, PuckettJW, et al. (2012) Role of mismatch repair enzymes in GAA.TTC triplet-repeat expansion in Friedreich ataxia induced pluripotent stem cells. J Biol Chem 287: 29861–29872.

104. EzzatizadehV, PintoRM, SandiC, SandiM, Al-MahdawiS, et al. (2012) The mismatch repair system protects against intergenerational GAA repeat instability in a Friedreich ataxia mouse model. Neurobiol Dis 46: 165–171.

105. KimHM, NarayananV, MieczkowskiPA, PetesTD, KrasilnikovaMM, et al. (2008) Chromosome fragility at GAA tracts in yeast depends on repeat orientation and requires mismatch repair. Embo J 27: 2896–2906.

106. KuS, SoragniE, CampauE, ThomasEA, AltunG, et al. (2010) Friedreich's ataxia induced pluripotent stem cells model intergenerational GAATTC triplet repeat instability. Cell Stem Cell 7: 631–637.

107. LiJL, HaydenMR, AlmqvistEW, BrinkmanRR, DurrA, et al. (2003) A genome scan for modifiers of age at onset in Huntington disease: The HD MAPS study. Am J Hum Genet 73: 682–687.

108. WexlerNS, LorimerJ, PorterJ, GomezF, MoskowitzC, et al. (2004) Venezuelan kindreds reveal that genetic and environmental factors modulate Huntington's disease age of onset. Proc Natl Acad Sci U S A 101: 3498–3503.

109. CowinRM, BuiN, GrahamD, GreenJR, Yuva-PaylorLA, et al. (2012) Genetic background modulates behavioral impairments in R6/2 mice and suggests a role for dominant genetic modifiers in Huntington's disease pathogenesis. Mamm Genome 23: 367–377.

110. KinT, OnoY (2007) Idiographica: a general-purpose web application to build idiograms on-demand for human, mouse and rat. Bioinformatics 23: 2945–2946.

111. Gomes-PereiraM, BidichandaniSI, MoncktonDG (2004) Analysis of unstable triplet repeats using small-pool polymerase chain reaction. Methods Mol Biol 277: 61–76.

112. HsiehYC, SkacelNE, BansalN, ScottoKW, BanerjeeD, et al. (2009) Species-specific differences in translational regulation of dihydrofolate reductase. Mol Pharmacol 76: 723–733.

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

114. KatohK, MisawaK, KumaK, MiyataT (2002) MAFFT: a novel method for rapid multiple sequence alignment based on fast Fourier transform. Nucleic Acids Res 30: 3059–3066.

115. BermanHM, WestbrookJ, FengZ, IypeL, SchneiderB, et al. (2003) The nucleic acid database. Methods Biochem Anal 44: 199–216.

116. KrivovGG, ShapovalovMV, DunbrackRLJr (2009) Improved prediction of protein side-chain conformations with SCWRL4. Proteins 77: 778–795.

117. KabschW, SanderC (1983) Dictionary of protein secondary structure: pattern recognition of hydrogen-bonded and geometrical features. Biopolymers 22: 2577–2637.

118. VenkatachalamCM (1968) Stereochemical criteria for polypeptides and proteins. VI. Non-bonded energy of polyglycine and poly-L-alanine in the crystalline beta-form. Biochim Biophys Acta 168: 411–416.

119. HutchinsonEG, ThorntonJM (1996) PROMOTIF–a program to identify and analyze structural motifs in proteins. Protein Sci 5: 212–220.

120. ClampM, CuffJ, SearleSM, BartonGJ (2004) The Jalview Java alignment editor. Bioinformatics 20: 426–427.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 2
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#