#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Genome-Wide Analysis Points to Roles for Extracellular Matrix Remodeling, the Visual Cycle, and Neuronal Development in Myopia


Myopia, or nearsightedness, is the most common eye disorder, resulting primarily from excess elongation of the eye. The etiology of myopia, although known to be complex, is poorly understood. Here we report the largest ever genome-wide association study (45,771 participants) on myopia in Europeans. We performed a survival analysis on age of myopia onset and identified 22 significant associations (), two of which are replications of earlier associations with refractive error. Ten of the 20 novel associations identified replicate in a separate cohort of 8,323 participants who reported if they had developed myopia before age 10. These 22 associations in total explain 2.9% of the variance in myopia age of onset and point toward a number of different mechanisms behind the development of myopia. One association is in the gene PRSS56, which has previously been linked to abnormally small eyes; one is in a gene that forms part of the extracellular matrix (LAMA2); two are in or near genes involved in the regeneration of 11-cis-retinal (RGR and RDH5); two are near genes known to be involved in the growth and guidance of retinal ganglion cells (ZIC2, SFRP1); and five are in or near genes involved in neuronal signaling or development. These novel findings point toward multiple genetic factors involved in the development of myopia and suggest that complex interactions between extracellular matrix remodeling, neuronal development, and visual signals from the retina may underlie the development of myopia in humans.


Vyšlo v časopise: Genome-Wide Analysis Points to Roles for Extracellular Matrix Remodeling, the Visual Cycle, and Neuronal Development in Myopia. PLoS Genet 9(2): e32767. doi:10.1371/journal.pgen.1003299
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003299

Souhrn

Myopia, or nearsightedness, is the most common eye disorder, resulting primarily from excess elongation of the eye. The etiology of myopia, although known to be complex, is poorly understood. Here we report the largest ever genome-wide association study (45,771 participants) on myopia in Europeans. We performed a survival analysis on age of myopia onset and identified 22 significant associations (), two of which are replications of earlier associations with refractive error. Ten of the 20 novel associations identified replicate in a separate cohort of 8,323 participants who reported if they had developed myopia before age 10. These 22 associations in total explain 2.9% of the variance in myopia age of onset and point toward a number of different mechanisms behind the development of myopia. One association is in the gene PRSS56, which has previously been linked to abnormally small eyes; one is in a gene that forms part of the extracellular matrix (LAMA2); two are in or near genes involved in the regeneration of 11-cis-retinal (RGR and RDH5); two are near genes known to be involved in the growth and guidance of retinal ganglion cells (ZIC2, SFRP1); and five are in or near genes involved in neuronal signaling or development. These novel findings point toward multiple genetic factors involved in the development of myopia and suggest that complex interactions between extracellular matrix remodeling, neuronal development, and visual signals from the retina may underlie the development of myopia in humans.


Zdroje

1. VitaleS, SperdutoRD, FerrisFL (2009) Increased prevalence of myopia in the United States between 1971–1972 and 1999–2004. Arch Ophthalmol 127: 1632–1639.

2. KempenJH, MitchellP, LeeKE, TielschJM, BromanAT, et al. (2004) The prevalence of refractive errors among adults in the United States, Western Europe, and Australia. Arch Ophthalmol 122: 495–505.

3. WojciechowskiR (2011) Nature and nurture: the complex genetics of myopia and refractive error. Clin Genet 79: 301–320.

4. RymerJ, WildsoetCF (2005) The role of the retinal pigment epithelium in eye growth regulation and myopia: a review. Vis Neurosci 22: 251–261.

5. RadaJA, SheltonS, NortonTT (2006) The sclera and myopia. Exp Eye Res 82: 185–200.

6. WildsoetC, WallmanJ (1995) Choroidal and scleral mechanisms of compensation for spectacle lenses in chicks. Vision Res 35: 1175–1194.

7. MuttiDO, ZadnikK, AdamsAJ (1996) Myopia. The nature versus nurture debate goes on. Invest Ophthalmol Vis Sci 37: 952–957.

8. FledeliusHC (1982) Ophthalmic changes from age of 10 to 18 years. A longitudinal study of sequels to low birth weight. IV. Ultrasound oculometry of vitreous and axial length. Acta Ophthalmol (Copenh) 60: 403–411.

9. FanDS, LamDS, LamRF, LauJT, ChongKS, et al. (2004) Prevalence, incidence, and progression of myopia of school children in Hong Kong. Invest Ophthalmol Vis Sci 45: 1071–1075.

10. FledeliusHC (1995) Myopia of adult onset. Can analyses be based on patient memory? Acta Ophthalmol Scand 73: 394–396.

11. IribarrenR, CortinezMF, ChiappeJP (2009) Age of first distance prescription and final myopic refractive error. Ophthalmic Epidemiol 16: 84–89.

12. IribarrenR, CerrellaMR, ArmestoA, IribarrenG, FornaciariA (2004) Age of lens use onset in a myopic sample of office-workers. Curr Eye Res 28: 175–180.

13. IsazaG, AroraS (2012) Incidence and severity of retinopathy of prematurity in extremely premature infants. Can J Ophthalmol 47: 296–300.

14. KennedyRH, BourneWM, DyerJA (1986) A 48-year clinical and epidemiologic study of keratoconus. Am J Ophthalmol 101: 267–273.

15. RichardsAJ, McNinchA, MartinH, OakhillK, RaiH, et al. (2010) Stickler syndrome and the vitreous phenotype: mutations in COL2A1 and COL11A1. Hum Mutat 31: E1461–1471.

16. HammondCJ, SniederH, GilbertCE, SpectorTD (2001) Genes and environment in refractive error: the twin eye study. Invest Ophthalmol Vis Sci 42: 1232–1236.

17. LyhneN, SjolieAK, KyvikKO, GreenA (2001) The importance of genes and environment for ocular refraction and its determiners: a population based study among 20–45 year old twins. Br J Ophthalmol 85: 1470–1476.

18. TeikariJM, O'DonnellJ, KaprioJ, KoskenvuoM (1991) Impact of heredity in myopia. Hum Hered 41: 151–156.

19. WojciechowskiR, CongdonN, BowieH, MunozB, GilbertD, et al. (2005) Heritability of refractive error and familial aggregation of myopia in an elderly American population. Invest Ophthalmol Vis Sci 46: 1588–1592.

20. PeetJA, CotchMF, WojciechowskiR, Bailey-WilsonJE, StambolianD (2007) Heritability and familial aggregation of refractive error in the Old Order Amish. Invest Ophthalmol Vis Sci 48: 4002–4006.

21. GossDA, JacksonTW (1996) Clinical findings before the onset of myopia in youth: 4. Parental history of myopia. Optom Vis Sci 73: 279–282.

22. KleinAP, DuggalP, LeeKE, KleinR, Bailey-WilsonJE, et al. (2005) Support for polygenic influences on ocular refractive error. Invest Ophthalmol Vis Sci 46: 442–446.

23. AshtonGC (1985) Segregation analysis of ocular refraction and myopia. Hum Hered 35: 232–239.

24. SoloukiAM, VerhoevenVJ, van DuijnCM, VerkerkAJ, IkramMK, et al. (2010) A genome-wide association study identifies a susceptibility locus for refractive errors and myopia at 15q14. Nat Genet 42: 897–901.

25. HysiPG, YoungTL, MackeyDA, AndrewT, Fernandez-MedardeA, et al. (2010) A genome-wide association study for myopia and refractive error identifies a susceptibility locus at 15q25. Nat Genet 42: 902–905.

26. LiYJ, GohL, KhorCC, FanQ, YuM, et al. (2011) Genome-wide association studies reveal genetic variants in CTNND2 for high myopia in Singapore Chinese. Ophthalmology 118: 368–375.

27. NakanishiH, YamadaR, GotohN, HayashiH, YamashiroK, et al. (2009) A genome-wide association analysis identified a novel susceptible locus for pathological myopia at 11q24.1. PLoS Genet 5: e1000660.

28. LiZ, QuJ, XuX, ZhouX, ZouH, et al. (2011) A genome-wide association study reveals association between common variants in an intergenic region of 4q25 and high-grade myopia in the Chinese Han population. Hum Mol Genet 20: 2861–2868.

29. ShiY, QuJ, ZhangD, ZhaoP, ZhangQ, et al. (2011) Genetic variants at 13q12.12 are associated with high myopia in the Han Chinese population. Am J Hum Genet 88: 805–813.

30. FanQ, BarathiVA, ChengCY, ZhouX, MeguroA, et al. (2012) Genetic variants on chromosome 1q41 influence ocular axial length and high myopia. PLoS Genet 8: e1002753.

31. YoungTL, MetlapallyR, ShayAE (2007) Complex trait genetics of refractive error. Arch Ophthalmol 125: 38–48.

32. ShiY, LiY, ZhangD, ZhangH, LiY, et al. (2011) Exome sequencing identifies ZNF644 mutations in high myopia. PLoS Genet 7: e1002084.

33. VerhoevenVJ, HysiPG, SawSM, VitartV, MirshahiA, et al. (2012) Large scale international replication and meta-analysis study confirms association of the 15q14 locus with myopia. The CREAM consortium. Hum Genet 131: 1467–1480.

34. PaluruPC, NallasamyS, DevotoM, RappaportEF, YoungTL (2005) Identification of a novel locus on 2q for autosomal dominant high-grade myopia. Invest Ophthalmol Vis Sci 46: 2300–2307.

35. YangZ, XiaoX, LiS, ZhangQ (2009) Clinical and linkage study on a consanguineous Chinese family with autosomal recessive high myopia. Mol Vis 15: 312–318.

36. NallasamyS, PaluruPC, DevotoM, WassermanNF, ZhouJ, et al. (2007) Genetic linkage study of high-grade myopia in a Hutterite population from South Dakota. Mol Vis 13: 229–236.

37. PaluruP, RonanSM, HeonE, DevotoM, WildenbergSC, et al. (2003) New locus for autosomal dominant high myopia maps to the long arm of chromosome 17. Invest Ophthalmol Vis Sci 44: 1830–1836.

38. BystromB, VirtanenI, RousselleP, GullbergD, Pedrosa-DomellofF (2006) Distribution of laminins in the developing human eye. Invest Ophthalmol Vis Sci 47: 777–785.

39. PetersonPE, PowCS, WilsonDB, HendrickxAG (1995) Localisation of glycoproteins and glycosaminoglycans during early eye development in the macaque. J Anat 186 (Pt 1) 31–42.

40. MorissetteN, CarbonettoS (1995) Laminin alpha 2 chain (M chain) is found within the pathway of avian and murine retinal projections. J Neurosci 15: 8067–8082.

41. BellSE, MavilaA, SalazarR, BaylessKJ, KanagalaS, et al. (2001) Differential gene expression during capillary morphogenesis in 3D collagen matrices: regulated expression of genes involved in basement membrane matrix assembly, cell cycle progression, cellular differentiation and G-protein signaling. J Cell Sci 114: 2755–2773.

42. VeyrierasJB, KudaravalliS, KimSY, DermitzakisET, GiladY, et al. (2008) High-resolution mapping of expression-QTLs yields insight into human gene regulation. PLoS Genet 4: e1000214.

43. StrangerBE, NicaAC, ForrestMS, DimasA, BirdCP, et al. (2007) Population genomics of human gene expression. Nat Genet 39: 1217–1224.

44. BadisG, BergerMF, PhilippakisAA, TalukderS, GehrkeAR, et al. (2009) Diversity and complexity in DNA recognition by transcription factors. Science 324: 1720–1723.

45. BoyleA, HongE, HariharanM, ChengY, ShaubM, et al. (2012) Annotation of functional variation in personal genomes using RegulomeDB. Genome Res 22: 1790–1797.

46. StraussO (2005) The retinal pigment epithelium in visual function. Physiol Rev 85: 845–881.

47. WangNK, ChuangLH, LaiCC, ChouCL, ChuHY, et al. (2012) Multimodal fundus imaging in fundus albipunctatus with RDH5 mutation: a newly identified compound heterozygous mutation and review of the literature. Doc Ophthalmol 125: 51–62.

48. NakamuraM, HottaY, TanikawaA, TerasakiH, MiyakeY (2000) A high association with cone dystrophy in Fundus albipunctatus caused by mutations of the RDH5 gene. Invest Ophthalmol Vis Sci 41: 3925–3932.

49. WangQ, ChenQ, ZhaoK, WangL, WangL, et al. (2001) Update on the molecular genetics of retinitis pigmentosa. Ophthalmic Genet 22: 133–154.

50. BernalS, CalafM, Garcia-HoyosM, Garcia-SandovalB, RosellJ, et al. (2003) Study of the involvement of the RGR, CRPB1, and CRB1 genes in the pathogenesis of autosomal recessive retinitis pigmentosa. J Med Genet 40: e89.

51. PattnaikBR, HughesBA (2012) Effects of KCNQ channel modulators on the M-type potassium current in primate retinal pigment epithelium. Am J Physiol, Cell Physiol 302: C821–833.

52. ZhangX, YangD, HughesBA (2011) KCNQ5/K(v)7.5 potassium channel expression and subcellular localization in primate retinal pigment epithelium and neural retina. Am J Physiol, Cell Physiol 301: C1017–1026.

53. NairKS, Hmani-AifaM, AliZ, KearneyAL, Ben SalemS, et al. (2011) Alteration of the serine protease PRSS56 causes angle-closure glaucoma in mice and posterior microphthalmia in humans and mice. Nat Genet 43: 579–584.

54. OrrA, DubeMP, ZentenoJC, JiangH, AsselinG, et al. (2011) Mutations in a novel serine protease PRSS56 in families with nanophthalmos. Mol Vis 17: 1850–1861.

55. GalA, RauI, El MatriL, KreienkampHJ, FehrS, et al. (2011) Autosomal-recessive posterior microphthalmos is caused by mutations in PRSS56, a gene encoding a trypsin-like serine protease. Am J Hum Genet 88: 382–390.

56. ReisLM, TylerRC, SchilterKF, Abdul-RahmanO, InnisJW, et al. (2011) BMP4 loss-of-function mutations in developmental eye disorders including SHORT syndrome. Hum Genet 130: 495–504.

57. BakraniaP, EfthymiouM, KleinJC, SaltA, BunyanDJ, et al. (2008) Mutations in BMP4 cause eye, brain, and digit developmental anomalies: overlap between the BMP4 and hedgehog signaling pathways. Am J Hum Genet 82: 304–319.

58. LipskaBS, BrzeskwiniewiczM, WierzbaJ, MorzuchiL, PiotrowskiA, et al. (2011) 8.6 Mb interstitial deletion of chromosome 4q13.3q21.23 in a boy with cognitive impairment, short stature, hearing loss, skeletal abnormalities and facial dysmorphism. Genet Couns 22: 353–363.

59. SchoenebeckJJ, HutchinsonSA, ByersA, BealeHC, CarringtonB, et al. (2012) Variation of BMP3 contributes to dog breed skull diversity. PLoS Genet 8: e1002849.

60. ScottSG, JunAS, ChakravartiS (2011) Sphere formation from corneal keratocytes and phenotype specific markers. Exp Eye Res 93: 898–905.

61. GudbjartssonDF, WaltersGB, ThorleifssonG, StefanssonH, HalldorssonBV, et al. (2008) Many sequence variants affecting diversity of adult human height. Nat Genet 40: 609–615.

62. LapanSW, ReddienPW (2011) dlx and sp6-9 Control optic cup regeneration in a prototypic eye. PLoS Genet 7: e1002226.

63. StoykovaA, FritschR, WaltherC, GrussP (1996) Forebrain patterning defects in Small eye mutant mice. Development 122: 3453–3465.

64. Garcia-FrigolaC, CarreresMI, VegarC, MasonC, HerreraE (2008) Zic2 promotes axonal divergence at the optic chiasm midline by EphB1-dependent and -independent mechanisms. Development 135: 1833–1841.

65. HerreraE, BrownL, ArugaJ, RachelRA, DolenG, et al. (2003) Zic2 patterns binocular vision by specifying the uncrossed retinal projection. Cell 114: 545–557.

66. Garcia-FrigolaC, HerreraE (2010) Zic2 regulates the expression of Sert to modulate eye-specific refinement at the visual targets. EMBO J 29: 3170–3183.

67. EsteveP, SandonisA, IbanezC, ShimonoA, GuerreroI, et al. (2011) Secreted frizzled-related proteins are required for Wnt/-catenin signalling activation in the vertebrate optic cup. Development 138: 4179–4184.

68. EsteveP, SandonisA, CardozoM, MalapeiraJ, IbanezC, et al. (2011) SFRPs act as negative modulators of ADAM10 to regulate retinal neurogenesis. Nat Neurosci 14: 562–569.

69. Garcia-HoyosM, CantalapiedraD, ArroyoC, EsteveP, RodriguezJ, et al. (2004) Evaluation of SFRP1 as a candidate for human retinal dystrophies. Mol Vis 10: 426–431.

70. EsteveP, TrousseF, RodriguezJ, BovolentaP (2003) SFRP1 modulates retina cell differentiation through a beta-catenin-independent mechanism. J Cell Sci 116: 2471–2481.

71. RodriguezJ, EsteveP, WeinlC, RuizJM, FerminY, et al. (2005) SFRP1 regulates the growth of retinal ganglion cell axons through the Fz2 receptor. Nat Neurosci 8: 1301–1309.

72. FogelBL, WexlerE, WahnichA, FriedrichT, VijayendranC, et al. (2012) RBFOX1 regulates both splicing and transcriptional networks in human neuronal development. Hum Mol Genet 21: 4171–4186.

73. LinJC, HoWH, GurneyA, RosenthalA (2003) The netrin-G1 ligand NGL-1 promotes the outgrowth of thalamocortical axons. Nat Neurosci 6: 1270–1276.

74. OlivaC, EscobedoP, AstorgaC, MolinaC, SierraltaJ (2012) Role of the MAGUK protein family in synapse formation and function. Dev Neurobiol 72: 57–72.

75. WalshT, PierceSB, LenzDR, BrownsteinZ, Dagan-RosenfeldO, et al. (2010) Genomic duplication and overexpression of TJP2/ZO-2 leads to altered expression of apoptosis genes in progressive nonsyndromic hearing loss DFNA51. Am J Hum Genet 87: 101–109.

76. ErikssonN, MacphersonJM, TungJY, HonLS, NaughtonB, et al. (2010) Web-based, participant-driven studies yield novel genetic associations for common traits. PLoS Genet 6: e1000993.

77. TungJY, DoCB, HindsDA, KieferAK, MacphersonJM, et al. (2011) Efficient Replication of over 180 Genetic Associations with Self-Reported Medical Data. PLoS ONE 6: e23473.

78. ErikssonN, BentonGM, DoCB, KieferAK, MountainJL, et al. (2012) Genetic variants associated with breast size also influence breast cancer risk. BMC Med Genet 13: 53.

79. ErikssonN, TungJY, KieferAK, HindsDA, FranckeU, et al. (2012) Novel associations for hypothyroidism include known autoimmune risk loci. PLoS ONE 7: e34442.

80. HennB, HonL, MacphersonJM, ErikssonN, SaxonovS, et al. (2012) Cryptic distant relatives are common in both isolated and cosmopolitan genetic samples. PLoS ONE 7: e34267.

81. Therneau T (2012) A Package for Survival Analysis in S. R package version 2.36-14.

82. SchemperM, StareJ (1996) Explained variation in survival analysis. Stat Med 15: 1999–2012.

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#