#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

The Impact of Population Demography and Selection on the Genetic Architecture of Complex Traits


Many human populations have dramatically expanded over the last several thousand years. I use population genetic models to investigate how recent population expansions affect patterns of mutations that reduce reproductive fitness and contribute to the genetic basis of complex traits (including common disease). I show that recent population growth increases the proportion of mutations found in the population that reduce fitness. When mutations that have the greatest effect on reproductive fitness also have the greatest effect on a complex trait, more of the heritability of the trait is due to mutations at very low-frequency in populations that have recently expanded, as compared to populations that have not. Also, under this model, for a given sample size and false-positive rate, fewer variants show statistically significant associations with the trait in the population that has expanded than in one that has not. Both of these findings suggest that recent population growth may make it more difficult to fully elucidate the genetic basis of complex traits that are directly or indirectly correlated with reproductive fitness.


Vyšlo v časopise: The Impact of Population Demography and Selection on the Genetic Architecture of Complex Traits. PLoS Genet 10(5): e32767. doi:10.1371/journal.pgen.1004379
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004379

Souhrn

Many human populations have dramatically expanded over the last several thousand years. I use population genetic models to investigate how recent population expansions affect patterns of mutations that reduce reproductive fitness and contribute to the genetic basis of complex traits (including common disease). I show that recent population growth increases the proportion of mutations found in the population that reduce fitness. When mutations that have the greatest effect on reproductive fitness also have the greatest effect on a complex trait, more of the heritability of the trait is due to mutations at very low-frequency in populations that have recently expanded, as compared to populations that have not. Also, under this model, for a given sample size and false-positive rate, fewer variants show statistically significant associations with the trait in the population that has expanded than in one that has not. Both of these findings suggest that recent population growth may make it more difficult to fully elucidate the genetic basis of complex traits that are directly or indirectly correlated with reproductive fitness.


Zdroje

1. AltshulerD, DalyMJ, LanderES (2008) Genetic mapping in human disease. Science 322: 881–888.

2. StrangerBE, StahlEA, RajT (2011) Progress and promise of genome-wide association studies for human complex trait genetics. Genetics 187: 367–383.

3. ManolioTA, CollinsFS, CoxNJ, GoldsteinDB, HindorffLA, et al. (2009) Finding the missing heritability of complex diseases. Nature 461: 747–753.

4. GibsonG (2012) Rare and common variants: Twenty arguments. Nat Rev Genet 13: 135–145.

5. EE, FlintJ, GibsonG, KongA, LealSM, et al. (2010) Missing heritability and strategies for finding the underlying causes of complex disease. Nat Rev Genet 11: 446–450.

6. NgSB, TurnerEH, RobertsonPD, FlygareSD, BighamAW, et al. (2009) Targeted capture and massively parallel sequencing of 12 human exomes. Nature 461: 272–276.

7. ShendureJ, JiH (2008) Next-generation DNA sequencing. Nat Biotechnol 26: 1135–1145.

8. WuMC, LeeS, CaiT, LiY, BoehnkeM, et al. (2011) Rare-variant association testing for sequencing data with the sequence kernel association test. Am J Hum Genet 89: 82–93.

9. MadsenBE, BrowningSR (2009) A groupwise association test for rare mutations using a weighted sum statistic. PLoS Genet 5: e1000384.

10. B, LealSM (2008) Methods for detecting associations with rare variants for common diseases: Application to analysis of sequence data. Am J Hum Genet 83: 311–321.

11. LiB, LealSM (2009) Discovery of rare variants via sequencing: Implications for the design of complex trait association studies. PLoS Genet 5: e1000481.

12. BamshadMJ, NgSB, BighamAW, TaborHK, EmondMJ, et al. (2011) Exome sequencing as a tool for Mendelian disease gene discovery. Nat Rev Genet 12: 745–755.

13. BamshadMJ, ShendureJA, ValleD, HamoshA, LupskiJR, et al. (2012) The centers for Mendelian genomics: A new large-scale initiative to identify the genes underlying rare Mendelian conditions. Am J Med Genet A 158A: 1523–1525.

14. NgSB, BighamAW, BuckinghamKJ, HannibalMC, McMillinMJ, et al. (2010) Exome sequencing identifies MLL2 mutations as a cause of Kabuki syndrome. Nat Genet 42: 790–793.

15. NgSB, BuckinghamKJ, LeeC, BighamAW, TaborHK, et al. (2010) Exome sequencing identifies the cause of a Mendelian disorder. Nat Genet 42: 30–35.

16. NgSB, NickersonDA, BamshadMJ, ShendureJ (2010) Massively parallel sequencing and rare disease. Hum Mol Genet 19: R119–24.

17. 1000 Genomes Project Consortium, Durbin RM, Abecasis GR, Altshuler DL, Auton A, et al (2010) A map of human genome variation from population-scale sequencing. Nature 467: 1061–1073.

18. 1000 Genomes Project Consortium, Abecasis GR, Auton A, Brooks LD, DePristo MA, et al (2012) An integrated map of genetic variation from 1,092 human genomes. Nature 491: 56–65.

19. CirulliET, GoldsteinDB (2010) Uncovering the roles of rare variants in common disease through whole-genome sequencing. Nat Rev Genet 11: 415–425.

20. GorlovIP, GorlovaOY, SunyaevSR, SpitzMR, AmosCI (2008) Shifting paradigm of association studies: Value of rare single-nucleotide polymorphisms. Am J Hum Genet 82: 100–112.

21. SchorkNJ, MurraySS, FrazerKA, TopolEJ (2009) Common vs. rare allele hypotheses for complex diseases. Curr Opin Genet Dev 19: 212–219.

22. KiezunA, GarimellaK, DoR, StitzielNO, NealeBM, et al. (2012) Exome sequencing and the genetic basis of complex traits. Nat Genet 44: 623–630.

23. HelgasonH, SulemP, DuvvariMR, LuoH, ThorleifssonG, et al. (2013) A rare nonsynonymous sequence variant in C3 is associated with high risk of age-related macular degeneration. Nat Genet 45: 1371–1374.

24. SeddonJM, YuY, MillerEC, ReynoldsR, TanPL, et al. (2013) Rare variants in CFI, C3 and C9 are associated with high risk of advanced age-related macular degeneration. Nat Genet 45: 1366–1370.

25. ZhanX, LarsonDE, WangC, KoboldtDC, SergeevYV, et al. (2013) Identification of a rare coding variant in complement 3 associated with age-related macular degeneration. Nat Genet 45: 1375–1379.

26. HeinzenEL, DepondtC, CavalleriGL, RuzzoEK, WalleyNM, et al. (2012) Exome sequencing followed by large-scale genotyping fails to identify single rare variants of large effect in idiopathic generalized epilepsy. Am J Hum Genet 91: 293–302.

27. NeedAC, McEvoyJP, GennarelliM, HeinzenEL, GeD, et al. (2012) Exome sequencing followed by large-scale genotyping suggests a limited role for moderately rare risk factors of strong effect in schizophrenia. Am J Hum Genet 91: 303–312.

28. HuntKA, MistryV, BockettNA, AhmadT, BanM, et al. (2013) Negligible impact of rare autoimmune-locus coding-region variants on missing heritability. Nature 498: 232–235.

29. LiuL, SaboA, NealeBM, NagaswamyU, StevensC, et al. (2013) Analysis of rare, exonic variation amongst subjects with autism spectrum disorders and population controls. PLoS Genet 9: e1003443.

30. LohmuellerKE, SparsoT, LiQ, AnderssonE, KorneliussenT, et al. (2013) Whole-exome sequencing of 2,000 Danish individuals and the role of rare coding variants in type 2 diabetes. Am J Hum Genet 93: 1072–1086.

31. TennessenJA, BighamAW, O'ConnorTD, FuW, KennyEE, et al. (2012) Evolution and functional impact of rare coding variation from deep sequencing of human exomes. Science 337: 64–69.

32. NelsonMR, WegmannD, EhmMG, KessnerD, St JeanP, et al. (2012) An abundance of rare functional variants in 202 drug target genes sequenced in 14,002 people. Science 337: 100–104.

33. FuW, O'ConnorTD, JunG, KangHM, AbecasisG, et al. (2013) Analysis of 6,515 exomes reveals the recent origin of most human protein-coding variants. Nature 493: 216–220.

34. MarthGT, YuF, IndapAR, GarimellaK, GravelS, et al. (2011) The functional spectrum of low-frequency coding variation. Genome Biol 12: R84–2011-12-9-r84.

35. CoventryA, Bull-OttersonLM, LiuX, ClarkAG, MaxwellTJ, et al. (2010) Deep resequencing reveals excess rare recent variants consistent with explosive population growth. Nat Commun 1: 131.

36. KeinanA, ClarkAG (2012) Recent explosive human population growth has resulted in an excess of rare genetic variants. Science 336: 740–743.

37. KryukovGV, ShpuntA, StamatoyannopoulosJA, SunyaevSR (2009) Power of deep, all-exon resequencing for discovery of human trait genes. Proc Natl Acad Sci U S A 106: 3871–3876.

38. SimonsYB, TurchinMC, PritchardJK, SellaG (2014) The deleterious mutation load is insensitive to recent population history. Nat Genet 46: 220–224.

39. WrightAF, CarothersAD, PirastuM (1999) Population choice in mapping genes for complex diseases. Nat Genet 23: 397–404.

40. PritchardJK, PrzeworskiM (2001) Linkage disequilibrium in humans: Models and data. Am J Hum Genet 69: 1–14.

41. PeltonenL, PalotieA, LangeK (2000) Use of population isolates for mapping complex traits. Nat Rev Genet 1: 182–190.

42. Service S, DeYoung J, Karayiorgou M, Roos JL, Pretorious H, et al (2006) Magnitude and distribution of linkage disequilibrium in population isolates and implications for genome-wide association studies. Nat Genet 38: 556–560.

43. ArdlieKG, KruglyakL, SeielstadM (2002) Patterns of linkage disequilibrium in the human genome. Nat Rev Genet 3: 299–309.

44. ReichDE, CargillM, BolkS, IrelandJ, SabetiPC, et al. (2001) Linkage disequilibrium in the human genome. Nature 411: 199–204.

45. AkeyJM, EberleMA, RiederMJ, CarlsonCS, ShriverMD, et al. (2004) Population history and natural selection shape patterns of genetic variation in 132 genes. PLoS Biol 2: e286.

46. BoykoAR, WilliamsonSH, IndapAR, DegenhardtJD, HernandezRD, et al. (2008) Assessing the evolutionary impact of amino acid mutations in the human genome. PLoS Genet 4: e1000083.

47. GutenkunstRN, HernandezRD, WilliamsonSH, BustamanteCD (2009) Inferring the joint demographic history of multiple populations from multidimensional SNP frequency data. PLoS Genet 5: e1000695.

48. KeinanA, MullikinJC, PattersonN, ReichD (2007) Measurement of the human allele frequency spectrum demonstrates greater genetic drift in East Asians than in Europeans. Nat Genet 39: 1251–1255.

49. LohmuellerKE, BustamanteCD, ClarkAG (2009) Methods for human demographic inference using haplotype patterns from genomewide single-nucleotide polymorphism data. Genetics 182: 217–231.

50. VoightBF, AdamsAM, FrisseLA, QianY, HudsonRR, et al. (2005) Interrogating multiple aspects of variation in a full resequencing data set to infer human population size changes. Proc Natl Acad Sci U S A 102: 18508–18513.

51. TennessenJA, O'ConnorTD, BamshadMJ, AkeyJM (2011) The promise and limitations of population exomics for human evolution studies. Genome Biol 12: 127.

52. GazaveE, MaL, ChangD, CoventryA, GaoF, et al. (2014) Neutral genomic regions refine models of recent rapid human population growth. Proc Natl Acad Sci U S A 111: 757–762.

53. LohmuellerKE, BustamanteCD, ClarkAG (2010) The effect of recent admixture on inference of ancient human population history. Genetics 185: 611–622.

54. LohmuellerKE, IndapAR, SchmidtS, BoykoAR, HernandezRD, et al. (2008) Proportionally more deleterious genetic variation in European than in African populations. Nature 451: 994–997.

55. SawyerSA, HartlDL (1992) Population genetics of polymorphism and divergence. Genetics 132: 1161–1176.

56. HernandezRD (2008) A flexible forward simulator for populations subject to selection and demography. Bioinformatics 24: 2786–2787.

57. HoggartCJ, Chadeau-HyamM, ClarkTG, LamparielloR, WhittakerJC, et al. (2007) Sequence-level population simulations over large genomic regions. Genetics 177: 1725–1731.

58. ClampM, FryB, KamalM, XieX, CuffJ, et al. (2007) Distinguishing protein-coding and noncoding genes in the human genome. Proc Natl Acad Sci U S A 104: 19428–19433.

59. Eyre-WalkerA (2010) Evolution in health and medicine Sackler Colloquium: Genetic architecture of a complex trait and its implications for fitness and genome-wide association studies. Proc Natl Acad Sci U S A 107 Suppl 11752–1756.

60. VisscherPM, HillWG, WrayNR (2008) Heritability in the genomics era—concepts and misconceptions. Nat Rev Genet 9: 255–266.

61. YangJ, LeeSH, GoddardME, VisscherPM (2011) GCTA: A tool for genome-wide complex trait analysis. Am J Hum Genet 88: 76–82.

62. ZaitlenN, KraftP (2012) Heritability in the genome-wide association era. Hum Genet 131: 1655–1664.

63. DempsterER, LernerIM (1950) Heritability of threshold characters. Genetics 35: 212–236.

64. RischNJ (2000) Searching for genetic determinants in the new millennium. Nature 405: 847–856.

65. GazaveE, ChangD, ClarkAG, KeinanA (2013) Population growth inflates the per-individual number of deleterious mutations and reduces their mean effect. Genetics 195: 969–978.

66. HaldaneJBS (1937) The effect of variation on fitness. Am Nat 71: 337–349.

67. MullerHJ (1950) Our load of mutations. Am J Hum Genet 2: 111–176.

68. PurcellSM, MoranJL, FromerM, RuderferD, SolovieffN, et al. (2014) A polygenic burden of rare disruptive mutations in schizophrenia. Nature 506: 185–190.

69. LangeLA, HuY, ZhangH, XueC, SchmidtEM, et al. (2014) Whole-exome sequencing identifies rare and low-frequency coding variants associated with LDL cholesterol. Am J Hum Genet 94: 233–245.

70. WrayNR, PurcellSM, VisscherPM (2011) Synthetic associations created by rare variants do not explain most GWAS results. PLoS Biol 9: e1000579.

71. VisscherPM, BrownMA, McCarthyMI, YangJ (2012) Five years of GWAS discovery. Am J Hum Genet 90: 7–24.

72. PritchardJK (2001) Are rare variants responsible for susceptibility to complex diseases? Am J Hum Genet 69: 124–137.

73. ThorntonKR, ForanAJ, LongAD (2013) Properties and modeling of GWAS when complex disease risk is due to non-complementing, deleterious mutations in genes of large effect. PLoS Genet 9: e1003258.

74. International Schizophrenia Consortium (2009) PurcellSM, WrayNR, StoneJL, VisscherPM, et al. (2009) Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature 460: 748–752.

75. Lango AllenH, EstradaK, LettreG, BerndtSI, WeedonMN, et al. (2010) Hundreds of variants clustered in genomic loci and biological pathways affect human height. Nature 467: 832–838.

76. WilliamsSM, HainesJL (2011) Correcting away the hidden heritability. Ann Hum Genet 75: 348–350.

77. KimuraM, MaruyamaT, CrowJF (1963) The mutation load in small populations. Genetics 48: 1303–1312.

78. RobertsDF, BillewiczWZ, McGregorIA (1978) Heritability of stature in a West African population. Ann Hum Genet 42: 15–24.

79. WrightA, CharlesworthB, RudanI, CarothersA, CampbellH (2003) A polygenic basis for late-onset disease. Trends Genet 19: 97–106.

80. SivakumaranS, AgakovF, TheodoratouE, PrendergastJG, ZgagaL, et al. (2011) Abundant pleiotropy in human complex diseases and traits. Am J Hum Genet 89: 607–618.

81. MaherMC, UricchioLH, TorgersonDG, HernandezRD (2012) Population genetics of rare variants and complex diseases. Hum Hered 74: 118–128.

82. PavardS, MetcalfCJ (2007) Negative selection on BRCA1 susceptibility alleles sheds light on the population genetics of late-onset diseases and aging theory. PLoS One 2: e1206.

83. LymanRF, LawrenceF, NuzhdinSV, MackayTF (1996) Effects of single P-element insertions on bristle number and viability in Drosophila melanogaster. Genetics 143: 277–292.

84. ParkJH, GailMH, WeinbergCR, CarrollRJ, ChungCC, et al. (2011) Distribution of allele frequencies and effect sizes and their interrelationships for common genetic susceptibility variants. Proc Natl Acad Sci U S A 108: 18026–18031.

85. TorgersonDG, BoykoAR, HernandezRD, IndapA, HuX, et al. (2009) Evolutionary processes acting on candidate cis-regulatory regions in humans inferred from patterns of polymorphism and divergence. PLoS Genet 5: e1000592.

86. Agarwala V, Flannick J, Sunyaev S, GoT2D Consortium, Altshuler D (2013) Evaluating empirical bounds on complex disease genetic architecture. Nat Genet 45: 1418–1427.

87. ZukO, SchaffnerSF, SamochaK, DoR, HechterE, et al. (2014) Searching for missing heritability: Designing rare variant association studies. Proc Natl Acad Sci U S A 111: E455–64.

88. DoR, KathiresanS, AbecasisGR (2012) Exome sequencing and complex disease: Practical aspects of rare variant association studies. Hum Mol Genet 21: R1–9.

89. KinnamonDD, HershbergerRE, MartinER (2012) Reconsidering association testing methods using single-variant test statistics as alternatives to pooling tests for sequence data with rare variants. PLoS One 7: e30238.

90. CastoAM, FeldmanMW (2011) Genome-wide association study SNPs in the human genome diversity project populations: Does selection affect unlinked SNPs with shared trait associations? PLoS Genet 7: e1001266.

91. ChenR, CoronaE, SikoraM, DudleyJT, MorganAA, et al. (2012) Type 2 diabetes risk alleles demonstrate extreme directional differentiation among human populations, compared to other diseases. PLoS Genet 8: e1002621.

92. BustamanteCD, BurchardEG, De la VegaFM (2011) Genomics for the world. Nature 475: 163–165.

93. CarlsonCS, MatiseTC, NorthKE, HaimanCA, FesinmeyerMD, et al. (2013) Generalization and dilution of association results from European GWAS in populations of non-European ancestry: The PAGE study. PLoS Biol 11: e1001661.

94. ShrinerD, AdeyemoA, GerryNP, HerbertA, ChenG, et al. (2009) Transferability and fine-mapping of genome-wide associated loci for adult height across human populations. PLoS One 4: e8398.

95. WatersKM, StramDO, HassaneinMT, Le MarchandL, WilkensLR, et al. (2010) Consistent association of type 2 diabetes risk variants found in Europeans in diverse racial and ethnic groups. PLoS Genet 6: e1001078.

96. MarigortaUM, NavarroA (2013) High trans-ethnic replicability of GWAS results implies common causal variants. PLoS Genet 9: e1003566.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 5
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#