#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Microsatellite Interruptions Stabilize Primate Genomes and Exist as Population-Specific Single Nucleotide Polymorphisms within Individual Human Genomes


Microsatellites are short tandem repeat DNA sequences located throughout the human genome that display a high degree of inter-individual variation. This characteristic makes microsatellites an attractive tool for population genetics and forensics research. Some microsatellites affect gene expression, and mutations within such microsatellites can cause disease. Interruption mutations disrupt the perfect repeated array and are frequently associated with altered disease risk, but they have not been thoroughly studied in human genomes. We identified interrupted mono-, di-, tri- and tetranucleotide MSs (iMS) within individual genomes from African, European, Asian and American population groups. We show that many iMSs, including some within disease-associated genes, are unique to a single population group. By measuring the conservation of microsatellites between human and chimpanzee genomes, we demonstrate that interruptions decrease the probability of microsatellite mutations throughout the genome. We demonstrate that iMSs arise in the human genome by single base changes within the DNA, and provide biochemical data suggesting that these stabilizing changes may be created by error-prone DNA polymerases. Our genome-wide study supports the model in which iMSs act to stabilize individual genomes, and suggests that population-specific differences in microsatellite architecture may be an avenue by which genetic ancestry impacts individual disease risk.


Vyšlo v časopise: Microsatellite Interruptions Stabilize Primate Genomes and Exist as Population-Specific Single Nucleotide Polymorphisms within Individual Human Genomes. PLoS Genet 10(7): e32767. doi:10.1371/journal.pgen.1004498
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004498

Souhrn

Microsatellites are short tandem repeat DNA sequences located throughout the human genome that display a high degree of inter-individual variation. This characteristic makes microsatellites an attractive tool for population genetics and forensics research. Some microsatellites affect gene expression, and mutations within such microsatellites can cause disease. Interruption mutations disrupt the perfect repeated array and are frequently associated with altered disease risk, but they have not been thoroughly studied in human genomes. We identified interrupted mono-, di-, tri- and tetranucleotide MSs (iMS) within individual genomes from African, European, Asian and American population groups. We show that many iMSs, including some within disease-associated genes, are unique to a single population group. By measuring the conservation of microsatellites between human and chimpanzee genomes, we demonstrate that interruptions decrease the probability of microsatellite mutations throughout the genome. We demonstrate that iMSs arise in the human genome by single base changes within the DNA, and provide biochemical data suggesting that these stabilizing changes may be created by error-prone DNA polymerases. Our genome-wide study supports the model in which iMSs act to stabilize individual genomes, and suggests that population-specific differences in microsatellite architecture may be an avenue by which genetic ancestry impacts individual disease risk.


Zdroje

1. LanderES, LintonLM, BirrenB, NusbaumC, ZodyMC, et al. (2001) Initial sequencing and analysis of the human genome. Nature 409: 860–921.

2. EllegrenH (2004) Microsatellites: simple sequences with complex evolution. Nat Rev Genet 5: 435–445.

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

4. LegendreM, PochetN, PakT, VerstrepenKJ (2007) Sequence-based estimation of minisatellite and microsatellite repeat variability. Genome Res 17: 1787–1796.

5. GemayelR, VincesMD, LegendreM, VerstrepenKJ (2010) Variable tandem repeats accelerate evolution of coding and regulatory sequences. Annu Rev Genet 44: 445–477.

6. HuiJ, HungLH, HeinerM, SchreinerS, NeumullerN, et al. (2005) Intronic CA-repeat and CA-rich elements: a new class of regulators of mammalian alternative splicing. EMBO J 24: 1988–1998.

7. LiYC, KorolAB, FahimaT, NevoE (2004) Microsatellites within genes: structure, function, and evolution. Mol Biol Evol 21: 991–1007.

8. KashiY, KingDG (2006) Simple sequence repeats as advantageous mutators in evolution. Trends Genet 22: 253–259.

9. RockmanMV, WrayGA (2002) Abundant raw material for cis-regulatory evolution in humans. Mol Biol Evol 19: 1991–2004.

10. NithianantharajahJ, HannanAJ (2007) Dynamic mutations as digital genetic modulators of brain development, function and dysfunction. BioEssays 29: 525–535.

11. FondonJW3rd, HammockEA, HannanAJ, KingDG (2008) Simple sequence repeats: genetic modulators of brain function and behavior. Trends Neurosci 31: 328–334.

12. HannanAJ (2010) Tandem repeat polymorphisms: modulators of disease susceptibility and candidates for ‘issing heritability’. Trends Genet 26: 59–65.

13. MatsuuraT, FangP, PearsonCE, JayakarP, AshizawaT, et al. (2006) Interruptions in the expanded ATTCT repeat of spinocerebellar ataxia type 10: repeat purity as a disease modifier? Am J Hum Genet 78: 125–129.

14. MatsuyamaZ, IzumiY, KameyamaM, KawakamiH, NakamuraS (1999) The effect of CAT trinucleotide interruptions on the age at onset of spinocerebellar ataxia type 1 (SCA1). J Med Genet 36: 546–548.

15. EichlerEE, HoldenJJ, PopovichBW, ReissAL, SnowK, et al. (1994) Length of uninterrupted CGG repeats determines instability in the FMR1 gene. Nat Genet 8: 88–94.

16. KunstCB, WarrenST (1994) Cryptic and polar variation of the fragile X repeat could result in predisposing normal alleles. Cell 77: 853–861.

17. BraidaC, StefanatosRKA, AdamB, MahajanN, SmeetsHJM, et al. (2010) Variant CCG and GGC repeats within the CTG expansion dramatically modify mutational dynamics and likely contribute toward unusual symptoms in some myotonic dystrophy type 1 patients. Human Mol Genet 19: 1399–1412.

18. LeeflangEP, ArnheimN (1995) A novel repeat structure at the myotonic dystrophy locus in a 37 repeat allele with unexpectedly high stability. Human MolGenet 4: 135–136.

19. LakenSJ, PetersenGM, GruberSB, OddouxC, OstrerH, et al. (1997) Familial colorectal cancer in Ashkenazim due to a hypermutable tract in APC. Nat Genet 17: 79–83.

20. YuZ, ZhuY, Chen-PlotkinAS, Clay-FalconeD, McCluskeyL, et al. (2011) PolyQ repeat expansions in ATXN2 associated with ALS are CAA interrupted repeats. PloS One 6: e17951.

21. RamosEM, MartinsS, AlonsoI, EmmelVE, Saraiva-PereiraML, et al. (2010) Common origin of pure and interrupted repeat expansions in spinocerebellar ataxia type 2 (SCA2). Am J Med Genet B Neuropsychiatr Genet 153B: 524–531.

22. BachinskiLL, CzernuszewiczT, RamagliLS, SuominenT, ShriverMD, et al. (2009) Premutation allele pool in myotonic dystrophy type 2. Neurology 72: 490–497.

23. BuschiazzoE, GemmellNJ (2006) The rise, fall and renaissance of microsatellites in eukaryotic genomes. Bioessays 28: 1040–1050.

24. KelkarYD, StrubczewskiN, HileSE, ChiaromonteF, EckertKA, et al. (2010) What is a microsatellite: a computational and experimental definition based upon repeat mutational behavior at A/T and GT/AC repeats. Genome Biol Evol 2: 620–635.

25. AnandaG, WalshE, JacobKD, KrasilnikovaM, EckertKA, et al. (2013) Distinct Mutational Behaviors Differentiate Short Tandem Repeats from Microsatellites in the Human Genome. Genome Biol Evol 5: 606–620.

26. BaptisteBA, AnandaG, StrubczewskiN, LutzkaninA, KhooSJ, et al. (2013) Mature microsatellites: mechanisms underlying dinucleotide microsatellite mutational biases in human cells. G3: Genes, Genomes, Genet (Bethesda) 3: 451–463.

27. KelkarYD, EckertKA, ChiaromonteF, MakovaKD (2011) A matter of life or death: how microsatellites emerge in and vanish from the human genome. Genome Res 21: 2038–2048.

28. TaylorJS, DurkinJM, BredenF (1999) The death of a microsatellite: a phylogenetic perspective on microsatellite interruptions. Mol Biol Evol 16: 567–572.

29. Reza ShariflouM, MoranC (2000) Conservation within artiodactyls of an AATA interrupt in the IGF-I microsatellite for 19–35 million years. Mol Biol Evol 17: 665–669.

30. EstoupA, TailliezC, CornuetJM, SolignacM (1995) Size homoplasy and mutational processes of interrupted microsatellites in two bee species, Apis mellifera and Bombus terrestris (Apidae). Mol Biol Evol 12: 1074–1084.

31. BrandstromM, EllegrenH (2008) Genome-wide analysis of microsatellite polymorphism in chicken circumventing the ascertainment bias. Genome Res 18: 881–887.

32. PetesTD, GreenwellPW, DominskaM (1997) Stabilization of microsatellite sequences by variant repeats in the yeast Saccharomyces cerevisiae. Genetics 146: 491–498.

33. RolfsmeierML, LahueRS (2000) Stabilizing effects of interruptions on trinucleotide repeat expansions in Saccharomyces cerevisiae. Mol Cell Biol 20: 173–180.

34. BrinkmannB, KlintscharM, NeuhuberF, HuhneJ, RolfB (1998) Mutation rate in human microsatellites: influence of the structure and length of the tandem repeat. Am J Hum Genet 62: 1408–1415.

35. WebsterMT, SmithNG, EllegrenH (2002) Microsatellite evolution inferred from human-chimpanzee genomic sequence alignments. Proc Natl Acad Sci USA 99: 8748–8753.

36. AbecasisGR, AutonA, BrooksLD, DePristoMA, DurbinRM, et al. (2012) An integrated map of genetic variation from 1,092 human genomes. Nature 491: 56–65.

37. MarthG, SchulerG, YehR, DavenportR, AgarwalaR, et al. (2003) Sequence variations in the public human genome data reflect a bottlenecked population history. Proc Natl Acad Sci USA 100: 376–381.

38. 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.

39. AmielJ, TrochetD, Clement-ZizaM, MunnichA, LyonnetS (2004) Polyalanine expansions in human. Human Mol Genet 13 Spec No 2: R235–243.

40. ChelalaC, KhanA, LemoineNR (2009) SNPnexus: a web database for functional annotation of newly discovered and public domain single nucleotide polymorphisms. Bioinformatics 25: 655–661.

41. Dayem UllahAZ, LemoineNR, ChelalaC (2012) SNPnexus: a web server for functional annotation of novel and publicly known genetic variants (2012 update). Nucl Acids Res 40: W65–70.

42. Dayem UllahAZ, LemoineNR, ChelalaC (2013) A practical guide for the functional annotation of genetic variations using SNPnexus. Brief Bioinform 14: 437–447.

43. GryfeR, Di NicolaN, LalG, GallingerS, RedstonM (1999) Inherited colorectal polyposis and cancer risk of the APC I1307K polymorphism. Am J Hum Genet 64: 378–384.

44. GryfeR, Di NicolaN, GallingerS, RedstonM (1998) Somatic instability of the APC I1307K allele in colorectal neoplasia. Cancer Res 58: 4040–4043.

45. EckertKA, MoweryA, HileSE (2002) Misalignment-mediated DNA polymerase beta mutations: comparison of microsatellite and frame-shift error rates using a forward mutation assay. Biochemistry 41: 10490–10498.

46. HileSE, WangX, LeeMY, EckertKA (2012) Beyond translesion synthesis: polymerase kappa fidelity as a potential determinant of microsatellite stability. Nucl Acids Res 40: 1636–1647.

47. EckertKA, HileSE, VargoPL (1997) Development and use of an in vitro HSV-tk forward mutation assay to study eukaryotic DNA polymerase processing of DNA alkyl lesions. Nucl Acids Res 25: 1450–1457.

48. BaptisteBA, EckertKA (2012) DNA polymerase kappa microsatellite synthesis: two distinct mechanisms of slippage-mediated errors. Environ Mol Mutagen 53: 787–796.

49. EckertKA, HileSE (2009) Every microsatellite is different: Intrinsic DNA features dictate mutagenesis of common microsatellites present in the human genome. Mol Carcinog 48: 379–388.

50. KelkarYD, TyekuchevaS, ChiaromonteF, MakovaKD (2008) The genome-wide determinants of human and chimpanzee microsatellite evolution. Genome Res 18: 30–38.

51. BebenekK, KunkelTA (2004) Functions of DNA polymerases. Adv Protein Chem 69: 137–165.

52. AbdulovicAL, HileSE, KunkelTA, EckertKA (2011) The in vitro fidelity of yeast DNA polymerase delta and polymerase varepsilon holoenzymes during dinucleotide microsatellite DNA synthesis. DNA Repair (Amst) 10: 497–505.

53. BrohedeJ, EllegrenH (1999) Microsatellite evolution: polarity of substitutions within repeats and neutrality of flanking sequences. Proc Biol Sci 266: 825–833.

54. VarelaMA, SanmiguelR, Gonzalez-TizonA, Martinez-LageA (2008) Heterogeneous nature and distribution of interruptions in dinucleotides may indicate the existence of biased substitutions underlying microsatellite evolution. J Mol Evol 66: 575–580.

55. BergstromTF, EngkvistH, ErlandssonR, JosefssonA, MackSJ, et al. (1999) Tracing the origin of HLA-DRB1 alleles by microsatellite polymorphism. Am J Hum Genet 64: 1709–1718.

56. PearsonCE, EichlerEE, LorenzettiD, KramerSF, ZoghbiHY, et al. (1998) Interruptions in the triplet repeats of SCA1 and FRAXA reduce the propensity and complexity of slipped strand DNA (S-DNA) formation. Biochemistry 37: 2701–2708.

57. JaremDA, HuckabyLV, DelaneyS (2010) AGG interruptions in (CGG)(n) DNA repeat tracts modulate the structure and thermodynamics of non-B conformations in vitro. Biochemistry 49: 6826–6837.

58. MadsenBE, VillesenP, WiufC (2008) Short tandem repeats in human exons: a target for disease mutations. BMC Genomics 9: 410.

59. DaiP, WongLJ (2003) Somatic instability of the DNA sequences encoding the polymorphic polyglutamine tract of the AIB1 gene. J Med Genet 40: 885–890.

60. GoodmanFR, MundlosS, MuragakiY, DonnaiD, Giovannucci-UzielliML, et al. (1997) Synpolydactyly phenotypes correlate with size of expansions in HOXD13 polyalanine tract. Proc Natl Acad Sci USA 94: 7458–7463.

61. EldenAC, KimHJ, HartMP, Chen-PlotkinAS, JohnsonBS, et al. (2010) Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 466: 1069–1075.

62. SobczakK, KrzyzosiakWJ (2004) Patterns of CAG repeat interruptions in SCA1 and SCA2 genes in relation to repeat instability. Hum Mutat 24: 236–247.

63. ChoudhryS, MukerjiM, SrivastavaAK, JainS, BrahmachariSK (2001) CAG repeat instability at SCA2 locus: anchoring CAA interruptions and linked single nucleotide polymorphisms. Human Mol Genet 10: 2437–2446.

64. MasutaniC, KusumotoR, YamadaA, DohmaeN, YokoiM, et al. (1999) The XPV (xeroderma pigmentosum variant) gene encodes human DNA polymerase eta. Nature 399: 700–704.

65. LinQ, ClarkAB, McCullochSD, YuanT, BronsonRT, et al. (2006) Increased susceptibility to UV-induced skin carcinogenesis in polymerase eta-deficient mice. Cancer Res 66: 87–94.

66. MasutaniC, KusumotoR, IwaiS, HanaokaF (2000) Mechanisms of accurate translesion synthesis by human DNA polymerase eta. EMBO J 19: 3100–3109.

67. ReyL, SidorovaJM, PugetN, BoudsocqF, BiardDS, et al. (2009) Human DNA polymerase eta is required for common fragile site stability during unperturbed DNA replication. Mol Cell Biol 29: 3344–3354.

68. BergoglioV, BoyerAS, WalshE, NaimV, LegubeG, et al. (2013) DNA synthesis by Pol eta promotes fragile site stability by preventing under-replicated DNA in mitosis. J Cell Biol 201: 395–408.

69. RogozinIB, PavlovYI, BebenekK, MatsudaT, KunkelTA (2001) Somatic mutation hotspots correlate with DNA polymerase eta error spectrum. Nat Immunol 2: 530–536.

70. ZengX, WinterDB, KasmerC, KraemerKH, LehmannAR, et al. (2001) DNA polymerase eta is an A-T mutator in somatic hypermutation of immunoglobulin variable genes. Nat Immunol 2: 537–541.

71. MasudaK, OuchidaR, HikidaM, KurosakiT, YokoiM, et al. (2007) DNA polymerases eta and theta function in the same genetic pathway to generate mutations at A/T during somatic hypermutation of Ig genes. J Biol Chem 282: 17387–17394.

72. PumpernikD, OblakB, BorstnikB (2008) Replication slippage versus point mutation rates in short tandem repeats of the human genome. Mol Genet Genomics 279: 53–61.

73. LoireE, HiguetD, NetterP, AchazG (2013) Evolution of coding microsatellites in primate genomes. Genome Biol Evol 5: 283–295.

74. HarrB, ZangerlB, SchlottererC (2000) Removal of microsatellite interruptions by DNA replication slippage: phylogenetic evidence from Drosophila. Mol Biol Evol 17: 1001–1009.

75. WalshE, WangX, LeeMY, EckertKA (2013) Mechanism of replicative DNA polymerase delta pausing and a potential role for DNA polymerase kappa in common fragile site replication. J Mol Biol 425: 232–243.

76. BetousR, PillaireMJ, PieriniL, van der LaanS, RecolinB, et al. (2013) DNA polymerase kappa-dependent DNA synthesis at stalled replication forks is important for CHK1 activation. EMBO J 32: 2172–2185.

77. Pena-DiazJ, BregenhornS, GhodgaonkarM, FollonierC, Artola-BoranM, et al. (2012) Noncanonical mismatch repair as a source of genomic instability in human cells. Mol Cell 47: 669–680.

78. Abajian C (1994–2003) Sputnik. Available: http://espressosoftware.com/sputnik/index.html.

79. LaiY, SunF (2003) The relationship between microsatellite slippage mutation rate and the number of repeat units. Mol Biol Evol 20: 2123–2131.

80. RheadB, KarolchikD, KuhnRM, HinrichsAS, ZweigAS, et al. (2010) The UCSC Genome Browser database: update 2010. Nucl Acids Res 38: D613–619.

81. KarolchikD, KuhnRM, BaertschR, BarberGP, ClawsonH, et al. (2008) The UCSC Genome Browser Database: 2008 update. Nucl Acids Res 36: D773–779.

82. KentWJ, SugnetCW, FureyTS, RoskinKM, PringleTH, et al. (2002) The human genome browser at UCSC. Genome Res 12: 996–1006.

83. HolsingerKE, WeirBS (2009) Genetics in geographically structured populations: defining, estimating and interpreting F(ST). Nat Rev Genet 10: 639–650.

84. JasjeetS (2011) Multivariate and Propensity Score Matching Software with Automated Balance Optimization: The Matching Package for R. J Statist Software 42: 1–52.

85. PurcellS, NealeB, Todd-BrownK, ThomasL, FerreiraMA, et al. (2007) PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 81: 559–575.

86. BlankenbergD, Von KusterG, CoraorN, AnandaG, LazarusR, et al. (2010) Galaxy: a web-based genome analysis tool for experimentalists. Curr Protoc Mol Biol Chapter 19: Unit 19 10 11–21.

87. GiardineB, RiemerC, HardisonRC, BurhansR, ElnitskiL, et al. (2005) Galaxy: a platform for interactive large-scale genome analysis. Genome Res 15: 1451–1455.

88. GoecksJ, NekrutenkoA, TaylorJ (2010) Galaxy: a comprehensive approach for supporting accessible, reproducible, and transparent computational research in the life sciences. Genome Biol 11: R86.

89. GrayKA, DaughertyLC, GordonSM, SealRL, WrightMW, et al. (2013) Genenames.org: the HGNC resources in 2013. Nucleic Acids Res 41 (Database issue) D545–52.

90. FalconS, GentlemanR (2007) Using GOstats to test gene lists for GO term association. Bioinformatics 23: 257–258.

91. OpreskoPL, ShimanR, EckertKA (2000) Hydrophobic interactions in the hinge domain of DNA polymerase beta are important but not sufficient for maintaining fidelity of DNA synthesis. Biochemistry 39: 11399–11407.

92. XieB, MazloumN, LiuL, RahmehA, LiH, et al. (2002) Reconstitution and characterization of the human DNA polymerase delta four-subunit holoenzyme. Biochemistry 41: 13133–13142.

93. HileSE, EckertKA (2008) DNA polymerase kappa produces interrupted mutations and displays polar pausing within mononucleotide microsatellite sequences. Nucl Acids Res 36: 688–696.

94. HileSE, EckertKA (2004) Positive correlation between DNA polymerase alpha-primase pausing and mutagenesis within polypyrimidine/polypurine microsatellite sequences. J Mol Biol 335: 745–759.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 7
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#