Seven new loci associated with age-related macular degeneration.
Journal: 2013/May - Nature Genetics
ISSN: 1546-1718
Abstract:
Age-related macular degeneration (AMD) is a common cause of blindness in older individuals. To accelerate the understanding of AMD biology and help design new therapies, we executed a collaborative genome-wide association study, including >17,100 advanced AMD cases and >60,000 controls of European and Asian ancestry. We identified 19 loci associated at P < 5 × 10(-8). These loci show enrichment for genes involved in the regulation of complement activity, lipid metabolism, extracellular matrix remodeling and angiogenesis. Our results include seven loci with associations reaching P < 5 × 10(-8) for the first time, near the genes COL8A1-FILIP1L, IER3-DDR1, SLC16A8, TGFBR1, RAD51B, ADAMTS9 and B3GALTL. A genetic risk score combining SNP genotypes from all loci showed similar ability to distinguish cases and controls in all samples examined. Our findings provide new directions for biological, genetic and therapeutic studies of AMD.
Relations:
Content
Citations
(250)
References
(61)
Grants
(1)
Clinical trials
(1)
Diseases
(1)
Conditions
(1)
Chemicals
(1)
Genes
(30)
Organisms
(1)
Processes
(2)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Nature genetics. Mar/31/2013; 45(4): 433-439e2
Published online Mar/2/2013

Seven New Loci Associated with Age-Related Macular Degeneration

Abstract

Age-related macular degeneration (AMD) is a common cause of blindness in older individuals. To accelerate understanding of AMD biology and help design new therapies, we executed a collaborative genomewide association study, examining >17,100 advanced AMD cases and >60,000 controls of European and Asian ancestry. We identified 19 genomic loci associated with AMD with p<5×10−8 and enriched for genes involved in regulation of complement activity, lipid metabolism, extracellular matrix remodeling and angiogenesis. Our results include 7 loci reaching p<5×10−8 for the first time, near the genes COL8A1/FILIP1L, IER3/DDR1, SLC16A8, TGFBR1, RAD51B, ADAMTS9/MIR548A2, and B3GALTL. A genetic risk score combining SNPs from all loci displayed similar good ability to distinguish cases and controls in all samples examined. Our findings provide new directions for biological, genetic and therapeutic studies of AMD.

AMD is a highly heritable progressive neurodegenerative disease that leads to loss of central vision through death of photoreceptors1,2. In developed countries, AMD is the leading cause of blindness in those >65 years3. Genes in the complement pathway411 and a region of chromosome 10 12,13 have now been implicated as the major genetic contributors to disease. Association has also been demonstrated with several additional loci1420, each providing an entry-point into AMD biology and potential therapeutic targets.

To accelerate the pace of discovery in macular degeneration genetics, 18 research groups from across the world formed the AMD Gene Consortium in early 2010, with support from the National Eye Institute (Table 1, Supplementary Table 1, Supplementary Note). To extend the catalog of disease associated common variants, we first organized a meta-analysis of genomewide association scans (GWAS) – combining data for >7,600 cases with advanced disease (geographic atrophy, neovascularization, or both) and >50,000 controls. Each study was first subject to GWAS quality control filters (customized taking into account study specific features21 as detailed in Supplementary Table 2) and standardized to the HapMap reference panel and statistical genotype imputation2225. Results were combined through meta-analysis26 and thirty-two variants representing loci with promising evidence of association were genotyped in an additional >9,500 cases and >8,200 controls (Supplementary Tables 1–3; summary meta-analysis results available online). Our overall analysis of the most promising variants thus included >17,100 cases and >60,000 controls.

Our meta-analysis evaluated evidence for association at 2,442,884 SNPs (Figure 1). Inspection of Q-Q plots (Supplementary Figure 1) and the genomic control value (λGC=1.06) suggest that unmodeled population stratification does not significantly impact our findings (Supplementary Table 4 for details). Joint analysis of discovery and follow-up studies27 resulted in 19 loci reaching p<5×10−8 (Figure 1, Table 2, Supplementary Table 5). These 19 loci include all susceptibility loci previously reaching p<5×10−8, except the 4q12 gene cluster for which association was reported in a Japanese population. In addition, the set includes seven loci reaching p<5×10−8 for the first time.

We evaluated heterogeneity between studies using the I2 statistic, which compares variability in effect size estimates between studies to chance expectations28. We observed significant (p<.05/19) heterogeneity only for loci near ARMS2 (I2=75.7%, p<1×10−6) and near CFH (I2=85.4%, p<1×10−6). Although these two loci were significantly associated in every sample examined, the magnitude of association varied more than expected. To explore sources of heterogeneity, we carried out a series of sub-analyses: we repeated the genomewide meta-analysis adding an age-adjustment, separating neovascular (NV) and geographic atrophy (GA) cases, in men and women, and in European- and Asian-ancestry samples separately (Figure 3, Supplementary Figure 2). These sub-analyses of the full GWAS dataset did not uncover additional loci reaching p<5×10−8; furthermore heterogeneity near CFH and ARMS2 remained significant in all sub-analyses (I2>65%, p <.001). Consistent with previous reports17,29,30, separate analysis of NV and GA cases showed ARMS2 risk alleles preferentially associated with risk of NV (ORNV=2.97, ORGA=2.50, pdifference=.0008) whereas CFH risk alleles preferentially associated with risk of GA (ORNV=2.34, ORGA=2.80, pdifference=.0006). We also observed large differences in effect sizes when stratifying by ethnicity, with variants near CFH exhibiting stronger evidence for association among Europeans (p=.0000001) and those near TNFRSF10A among East Asians (p=.002). Potential explanations include differences in linkage disequilibrium between populations or differences in environmental or diagnostic factors that modify genetic effects.

Identifying the full spectrum of allelic variation that contributes to disease in each locus will require sequencing of AMD cases and controls. To conduct an initial evaluation of the evidence for multiple AMD risk alleles in the nineteen loci described here, we repeated genomewide association analyses conditioning on the risk alleles listed in Table 2. We then examined each of the 19 implicated loci for variants with independent association (at p<.0002, corresponding for an estimate of ~250 independent variants per locus). This analysis resulted in the identification of the previously well documented independently associated variants near CFH and C2/CFB8,10,31,32 and of additional independent signals near C3, CETP, LIPC, FRK/COL10A1, IER3/DDR1, RAD51B (Supplementary Table 6). In four of these loci, the independently associated variants mapped very close (within <60kb) to the original signal. This shows each locus can harbor multiple susceptibility alleles, encouraging searches for rare variants that elucidate gene function in these regions33,34.

To prioritize our search for likely causal variants, we examined each locus in detail (see LocusZoom35 plots in Supplementary Figure 3) and investigated whether AMD risk alleles were associated with changes in protein sequence, copy number variation or insertion deletion polymorphisms. One quarter of associated variants altered protein sequence, either directly (N=2) or through linkage disequilibrium (r2>.6; N=3) with a nearby non-synonymous variant (Supplementary Table 7). Some coding variants point to well-studied genes (ARMS2, C3 and APOE) while others help prioritize nearby genes for further study. In chromosome 4q25, index SNP rs4698775 is in strong linkage disequilibrium (r2=.88) with a potentially protein damaging variant in CCDC109B36, a coiled coil domain containing protein that may be involved in the regulation of gene expression. In chromosome 6q22, index SNP rs3812111 is a perfect proxy for a coding variant in COL10A1, a collagen protein that could be important in maintaining the structure and function of the extra-cellular matrix. Interestingly, rs1061170 (NP_000177.2[CFH]:p.His402Tyr) was not in disequilibrium with rs10737680, the most strongly associated SNP in the CFH region but, instead, was tagged by a secondary and weaker association signal (Supplementary Tables 6&7). This is consistent with prior haplotype analyses of the locus10,31,32,34,37.

We used publicly available data38,39 to check whether any of our index SNPs might be proxies for copy number variants or insertion-deletion polymorphisms (indels), which are hard to directly interrogate with genotyping arrays. This analysis identified a single strong association (r2=.99), between rs10490924, a coding variant in the ARMS2 gene which is the peak of association in 10q26, and a 3′ UTR indel polymorphism associated with ARMS2 mRNA instability40. Since index SNP rs10490924 is also in strong disequilibrium (r2=.90) with a nearby SNP, rs11200638, that regulates HTRA141, our data does not directly answer whether HTRA1 or ARMS2 is the causal gene in this locus. Although a common deletion of the CFHR1 and CFHR3 genes has been proposed42,43, there was only modest signal in this study which is likely due to linkage disequilibrium with our most significantly associated variants in the locus (r2=.31 between rs10737680 and 1000 Genomes Project MERGED_DEL_2_6731) as previously suggested34.

Using RNA-sequencing44, we examined mRNA levels of 85 genes within 100 kb of our index SNPs in post-mortem human retina (Supplementary Table 8). Of 19 independent risk loci, three had no genes with expressed transcripts in either old or young retina. Two genes showed differential expression between post-mortem retina of young (ages 17–35) and elderly (ages 75 and 77) individuals: CFH (p=.009) and VEGFA (p=.003), both with increased expression in older individuals. Using previously published data45, we also examined the expression of associated genes in fetal and adult retinal pigment epithelium (RPE). This revealed increased C3 expression in adult RPE compared to fetal RPE (p=.0008). CFH, VEGFA and C3 are thus up-regulated with aging, and their role in AMD may indicate an accelerated aging process. In addition to C3 and CFH, all the complement genes with detectable expression in the retina or RPE experiments showed higher expression levels in older tissue.

To identify biological relationships among our genetic association signals, we catalogued the genes within 100kb of the variants in each association peak (r2>0.8 with the index SNP listed in Table 1). Ingenuity Pathway Analysis (Ingenuity Systems, Redwood, CA) highlighted several biological pathways, particularly the complement system and atherosclerotic signaling, to be enriched in the resulting set of 90 genes (Table 3, Supplementary Table 9). To account for features of genomewide association studies (such as the different number of SNPs in each gene), we carried out two additional analyses. First, we repeated our analysis for 50 sets of 19 control loci drawn from the NHGRI GWAS catalog46. In these 50 control sets, Ingenuity enrichment p-values for the complement system and for atherosclerosis signaling genes were exceeded 16% and 32% of the time respectively (although these two specific pathways were never implicated in a control dataset). We also repeated our enrichment analyses using the INRICH tool47, which is specifically designed for the analysis of genomewide association studies – but accesses a more limited set of annotations. The INRICH analyses showed enrichment for genes encoding collagen and extra-cellular region proteins (both with p=10−5 and after adjustment for multiple testing padjust=.0006), complement and coagulation cascades (p=.0002, padjust=.03), lipoprotein metabolism (p=.0003, padjust=.04), and regulation of apoptosis (p=.0009, padjust=.09) (Supplementary Table 10).

To explore the connections between our genetic association signals, we tested for interaction between pairs of risk alleles – looking for situations where joint risk was different than expected based on marginal effects. This analysis resulted in 171 tests of interaction, of which 9 were nominally significant (p<.05, see Supplementary Table 11), consistent with chance expectations. The strongest observed interaction involved risk alleles at rs10737680 (near CFH) and rs429608 (near C2/CFB), the only association that remained significant after adjusting for multiple testing (p=.000052, <0.05/171=.00029). Individuals carrying risk alleles at both these loci where at slightly higher risk of disease than expected.

The proportion of the variability in the risk of AMD that is due to genes, or heritability, has been estimated at 45–70% 2. Estimating the proportion of disease risk explained by the susceptibility loci identified 48 depends greatly on the disease prevalence - which is difficult to estimate in our sample, as it includes cases and controls of different ages and collected through a variety of ascertainment schemes. Using a model that assumes an underlying normally distributed but unobserved disease risk score or liability49, the nineteen loci described here account for between 10% (if AMD prevalence is close to 1%) and 30% (if AMD prevalence is closer to 10%) of the variability in disease risk (corresponding to 15–65% of the total genetic contribution to AMD). The variants representing the peak of association at loci previously reaching genomewide significance account for the bulk of this variability: the new loci identified here account for 0.5–1.0% of the total heritability of AMD whereas secondary signals at novel and known loci account for 1.5–3.0% of the total heritability.

We report here the most comprehensive genetic association study of macular degeneration yet conducted, involving 18 international research groups, and a large set of cases and controls. Our data reveal 19 susceptibility loci, including 7 loci reaching p<5×10−8 for the first time, nearly doubling the number of known AMD loci outside the complement pathway. Our results show some susceptibility alleles exhibit different association across ethnic groups and may be preferentially associated with specific subtypes of disease. As with other GWAS meta-analysis, differences in genotyping methods, quality control steps and imputation strategies between samples might have a minor effect in our results – future studies may document that more uniform approaches across larger sample sizes might uncover more signals. A conundrum of macular degeneration genetics remains that the loci identified to date contribute to both GA and NV, two different phenotypes of advanced disease. In our sample, subtype specific GWAS analyses considering GA or NV cases only did not identify additional loci. Consistent with observations for other complex diseases39, the majority of common disease susceptibility alleles do not alter protein sequences and are not associated with insertions or deletions of coding sequence or with copy number variation. We expect that the loci identified here will provide an ideal starting point for studies of rare variation33,34.

In contrast to most other complex diseases, a risk score combining information across our 19 loci, can distinguish cases and controls relatively well (Figure 4, area under the ROC curve [AUC]=.52 including only new loci or AUC=.74 including new and previously reported loci; Supplementary Figure 4). It may be possible to use similar scores to identify and prioritize at risk individuals so they receive preventative treatment prior to the onset of disease50. Monotherapies are increasingly utilized to manage neovascular disease, but offer only a limited repertoire of treatment options to patients. Identification of novel genes and pathways enables us to pursue a larger range of disease-specific targets for development of new therapeutic interventions. We expect that future therapies directed at earlier stages of the disease process will allow patients to retain visual function for longer periods, improving the quality of life for individuals with AMD.

ONLINE METHODS

GENOME-WIDE SCAN FOR LATE AMD ASSOCIATION INCLUDING FOLLOW-UP

Study-specific association analysis for discovery

Genotyping was performed on a variety of different platforms summarized in Supplementary Table 2. Each group submitted results from association tests using genotyped and imputed data where the allelic dosages were computed with either MACH25, IMPUTE23, BEAGLE24, or snpStats52 using the HapMap2 reference panels. The CEU panel was used as a reference for imputation-based analyses for most samples (predominantly of European ancestry), with two exceptions: for the JAREDS samples (predominantly of East Asian ancestry), the CHB+JPT panel was used as a reference; for the VRF samples (predominantly of South Asian ancestry) the combined CEU and CHB+JPT panels were used22,53. For most data sets association tests were run under a logistic regression model using either Plink54, Mach2dat25, ProbABEL55, or snpStats52, though for one dataset containing related individuals the generalized estimating equations algorithm56 as implemented in R57,58. In addition to the primary analysis which tested for SNP associations with advanced AMD unadjusted for age, an age-adjusted sensitivity analysis was conducted by each group with available age. Each group also provided stratified results by sex or AMD subtype (GA or NV) as long as the sample size per stratum exceeded 50 subjects. For all analyses, study-specific control for population stratification was conducted (Supplementary Table 4).

Study-specific association analysis for follow-up

Genotyping of the selected SNPs was performed on different platforms; the same models, sensitivity and stratified analyses were computed by each follow-up partner, while SNPs with insufficient call rate were excluded based on study-specific thresholds. If the index SNP could not be genotyped, a highly correlated proxy was used whenever possible (Supplementary Tables 2&3).

Quality control before meta-analysis

Before meta-analysis, all study-specific files underwent quality control procedures to check for completeness and plausible descriptive statistics on all variables as well as for compliance of allele frequencies with HapMap59. In addition, we excluded SNP results of a study into meta-analysis (i) for discovery: if imputation quality measures were too low (MACH & PLINK <0.3; SNPTEST <0.4) or if effect sizes (|beta|) or standard errors were too extreme (≥5) indicating instability of the estimates, (ii) for follow-up: if Hardy-Weinberg equilibrium was violated (p<0.05/32).

Meta-analyses

For both discovery and follow-up, we performed meta-analyses using the inverse variance weighted fixed effect model, which pools the effect size and standard error of each participated GWAS. Using an alternative weighted z-score method, which is based on a weighted sum of z-score statistics, we obtained a very similar set of test statistics (correlation of −log10(p-value) >0.98). All analyses were performed using METAL26 and R. For the discovery, we applied two rounds of genomic control corrections to each individual GWAS and the combined meta results, respectively 51. All results were analyzed and validated among four independent teams.

EXTENDED ANALYSES FOR THE IDENTIFIED AMD LOCI

Extended analyses were conducted on the identified loci and particularly on the top SNP of each locus.

Second signal analysis

To detect potential independent signals within the identified AMD loci, each study partner with genotypes for all identified SNPs available re-analyzed their data for all SNPs in the respective loci (index SNP ±1Mb) using a logistic regression model containing all identified index SNPs. Quality control procedures were performed as before. The beta estimates for each SNP were meta-analyzed applying the effective sample size weighted z-score method and two rounds of genomic control correction. The significance threshold (p<0.05) for an independent association signal within any of the identified loci was Bonferroni-adjusted using the average effective number of SNPs involved across the identified loci determined by SNPSpD60. To this analysis, 13 studies contributed including 7,489 cases and 51,562 controls.

Interaction analysis

Utilizing a pre-specified R-scripts (see URLs), GWAS partners performed 171 logistic regression analyses modeling the pair-wise interaction of the 19 index SNPs assuming an additive model for main and interaction effects. Study-specific covariates were included to the model if required. Per study, quality control included a check for consistency of SNP main effects between discovery and interaction analysis. SNPs with low imputation quality measures and pairs with |beta|>5 or standard errors >5 were excluded before meta-analyzing the interaction effects with the inverse variance weighted fixed effect model in METAL. To this analysis, 12 studies contributed including 6,645 cases and 49,410 controls.

GENETIC RISK SCORE

The meta-analyzed effect sizes, βj, for each of the 19 SNPs were calculated in the meta-analysis described above and normalized: β^j=βj/k=119βk, j=1,…,19. Using these as weights, each study partner with all 19 SNPs available computed the individuals’ genetic risk score as a normalized weighted sum of the AMD risk increasing alleles among the identified SNPs as

Si=jβ^jxij, where xij is the genotype of the ith individual at the jth SNP, so Si ranges from 0 to 2. This data was available from 12 studies including 7,195 cases and 49,149 controls.

For each study, we used a leave-one-out cross-validation to access the prediction of the risk score. For the kth subject, we fitted a logistic regression model from all subjects in the study excluding the kth subjects: log(yi1-yi)=α+γSi, i! = k, α is the intercept and γ is the effect of the genetic risk score. The fitted probability of the kth subject was then estimated as ŷk = 1/(1+e−(α̂+γ̂Sk)). We sorted the fitted probabilities and calculated sensitivity and specificity by varying the risk threshold (the value compared with the fitted probability to dichotomize the subject into case or control) from 0 to 1. These were utilized to compute the area-under-the curve (AUC) of the receiver-operating-curve (ROC).

IDENTIFICATION OF CORRELATED CODING VARIANTS AND TAGGED NON-SNP VARIATION

LD estimates were calculated using genotype data of the identified risk loci (index SNPs ±500kb) of individuals with European ancestry from the 1000 Genomes Project (March 2012 release)61 or from HapMap (release #28)59. Variants correlated (r²>0.6) with one of the GWAS index SNPs were identified using PLINK54. To filter coding variants, all correlated variants were mapped against RefSeq transcripts using ANNOVAR62.

GENE EXPRESSION

We evaluated expression of genes within 100kb of one of the 19 index SNPs, as well as of several retina-specific, RPE-specific and housekeeping genes unrelated to AMD for comparison in retina (RNA-sequencing data from three young [17–35 yrs age] and two old individuals [75 and 77 yrs age]) as well as in fetal and adult retinal pigment epithelium (RPE; published data in the Gene Expression Omnibus database45). Expression was analyzed using previously described protocols44 (Supplementary Table 8).

PATHWAY ANALYSES

Functional enrichment analysis was performed using the Ingenuity Pathway Analysis software (IPA, Ingenuity® Systems). Any gene located within 100kb of a SNP in high LD (r2>0.8) with one of the index SNPs was considered a potential AMD risk associated gene and considered for subsequent pathway enrichment analysis. LD estimates were calculated as described above. Applying the above inclusion filters, 90 genes appear to be implicated by our 19 replicated AMD SNPs (Supplementary Table 8). Because genes with related function sometimes cluster in the same locus, we trimmed gene lists during analysis so that only one gene per locus was used to evaluate enrichment for each pathway. The P-value of the association between our implicated gene list and any of the canonical pathways and/or functional gene sets as annotated by IPA’s Knowledge Base was computed using a one-sided Fisher’s exact test. The Benjamini-Hochberg method was used to estimate False Discovery Rates. To evaluate significance of observed enrichment, we repeated our Ingenuity analysis starting with 50 lists of 19 SNPs randomly drawn from the NHGRI GWAS catalog46 and, again, using the INRICH tool63. When using INRICH, we used gene sets defined in the Broad’s Molecular Signatures database47 (ver3.0) representing manually curated canonical pathway, Gene Ontology biological process, cellular component and molecular function gene sets (C2:CP, C5:BP, C5:CC and C5:MF). We provided INRICH with our full GWAS SNP list and allowed it to carry out 100,000 permutations, matching selected loci in terms of gene count, SNP density and total number of SNPs.

Supplementary Material

FIGURE 1

Summary of genomewide association scan results

Summary of genomewide association scan results in the discovery GWAS sample. Previously described loci reaching p < 5×10−8 are labeled in blue; new loci reaching p < 5×10−8 for the first time after follow-up are labeled in green.

FIGURE 2

Sensitivity analysis

The top left panel compares estimated effect sizes for the original analysis and for an age-adjusted analysis (where age was included as a covariate and samples of unknown age were excluded). The top right panel compares analyses stratified by sex. The bottom left panel evaluates stratification by disease subtype. The bottom right panel evaluates stratification by ethnicity. The size of each marker reflects confidence intervals (with height reflecting confidence interval along the Y axis and width reflecting confidence interval along the X axis). Comparisons reaching p < 0.05 are labeled and colored in red.

FIGURE 3

Risk score analysis

We calculated a risk score for each individual, defined as the product of the number of risk alleles at each locus and the associated effect size for each allele (measured on the log-odds scale). The plot summarizes the ability of these overall genetic risk scores to distinguish cases and controls.

TABLE 1

Summary of the Samples Used in Genomewide Discovery and Targeted Follow-Up Analyses

For additional details, including a breakdown of the number of cases and controls in individual samples, see Supplementary Table 1. NCASES includes only cases with geographic atrophy, choroidal neovasculartization, or both.

AnalysisContributing Study GroupsNCASES%Female%Neovascular DiseaseNCONTROLS% Female
Genomewide Discovery157,65053.959.251,84445.2
Targeted Follow-up189,53156.357.88,23053.8
Overall3317,18155.258.460,07446.3
TABLE 2

Summary of Loci Reaching Genome-Wide Significance

All results reported here include a genomic control correction for individual studies and also for the final meta-analysis51.

SNP/Risk AlleleChromosome, PositionNearby GenesEAFDiscoveryFollow-upJoint95% CI
PORPORPOR
Loci Previously Reported With P < 5×10−8
rs10490924/T10124.2 MbARMS2/HTRA10.304×10−3532.712.8×10−1902.884×10−5402.76[2.72–2.80]
rs10737680/A1196.7 MbCFH0.641×10−2832.402.7×10−1522.501×10−4342.43[2.39–2.47]
rs429608/G631.9 MbC2/CFB0.862×10−541.672.4×10−371.894×10−891.74[1.68–1.79]
rs2230199/C196.7 MbC30.202×10−261.463.4×10−171.371×10−411.42[1.37–1.47]
rs5749482/G2233.1 MbTIMP30.746×10−131.259.7×10−171.452×10−261.31[1.26–1.36]
rs4420638/A1945.4 MbAPOE0.833×10−151.344.2×10−71.252×10−201.30[1.24–1.36]
rs1864163/G1657 MbCETP0.768×10−131.258.7×10−51.177×10−161.22[1.17–1.27]
rs943080/T643.8 MbVEGFA0.514×10−121.181.6×10−51.129×10−161.15[1.12–1.18]
rs13278062/T823.1 MbTNFRSF10A0.487×10−101.176.4×10−71.143×10−151.15[1.12–1.19]
rs920915/C1558.7 MbLIPC0.482×10−91.140.0041.103×10−111.13[1.09–1.17]
rs4698775/G4110.6 MbCFI0.312×10−101.160.0251.087×10−111.14[1.10–1.17]
rs3812111/T6116.4 MbCOL10A10.647×10−81.130.0221.062×10−81.10[1.07–1.14]

Loci Reaching P < 5×10−8 for the First Time
rs13081855/T399.5 MbCOL8A1/FILIP1L0.104×10−111.286.0×10−41.174×10−131.23[1.17–1.29]
rs3130783/A630.8 MbIER3/DDR10.791×10−61.153.5×10−61.162×10−111.16[1.11–1.20]
rs8135665/T2238.5 MbSLC16A80.218×10−81.165.6×10−51.132×10−111.15[1.11–1.19]
rs334353/T9101.9 MbTGFBR10.739×10−71.136.7×10−61.133×10−111.13[1.10–1.17]
rs8017304/A1468.8 MbRAD51B0.619×10−71.112.1×10−51.119×10−111.11[1.08–1.14]
rs6795735/T364.7 MbADAMTS9/MIR548A20.469×10−81.130.00661.075×10−91.10[1.07–1.14]
rs9542236/C1331.8 MbB3GALTL0.442×10−61.120.00181.082×10−81.10[1.07–1.14]

See Supplementary Table 5 for a summary of all gene name abbreviations used in this Table and elsewhere in the paper. EAF is the allele frequency of the risk increasing allele.

TABLE 3
Pathway Analysis
Ingenuity Canonical PathwaysEnrichment Analysis
Nominal p-valueFDR q-valueMoleculesPathway Size(Ngenes)
Complement System0.0000120.0015CFI, CFH, C3, CFB*, C2*,C4A*, C4B*35
Atherosclerosis Signaling0.000140.009PLA2G12A, APOC1**, APOE**, APOC2**, APOC4**, TNFSF14, COL10A1, PLA2G6129
VEGF Family Ligand-Receptor Interactions0.00420.150VEGFA,PLA2G12A,PLA2G684
Dendritic Cell Maturation0.00460.150RELB, ZBTB12, DDR1, COL10A1185
Phospholipid Degradation0.00580.151PLA2G12A, LIPC, PLA2G6102
MIF-mediated Glucocorticoid Regulation0.00880.153PLA2G12A, PLA2G642
Inhibition of Angiogenesis by TSP10.00930.153VEGFA,TGFBR139
Fc Epsilon RI Signaling0.00980.153VAV1, PLA2G12A, PLA2G6111
p38 MAPK Signaling0.0110.153PLA2G12A, TGFBR1, PLA2G6106

*CFB, C2, C4A, and C4B all flank rs429608 and thus counted as single hit when determining significance of enrichment.

**APOC1, APOE, APOC2, and APOC4 all flank rs4420638 and thus counted as single hit when determining significance of enrichment.

Appendix

AUTHOR LIST

Lars G Fritsche [1,2, *], Wei Chen [2,3,*], Matthew Schu [4,*], Brian L Yaspan [5,6,*], Yi Yu [7,*], Gudmar Thorleifsson [8], Donald J Zack [9,10,11,12], Satoshi Arakawa [13], Valentina Cipriani [14], Stephan Ripke [15,16], Robert P Igo, Jr. [17,18], Gabriëlle H S Buitendijk [19,20], Xueling Sim [2,21], Daniel E Weeks [22,23], Robyn H Guymer [24], Joanna E Merriam [25], Peter J Francis [26], Gregory Hannum [27], Anita Agarwal [28,29], Ana Maria Armbrecht [30], Isabelle Audo [10,31,32,33], Tin Aung [34,35], Gaetano R Barile [25], Mustapha Benchaboune [36], Alan C Bird [14], Paul N Bishop [37,38], Kari E Branham [39], Matthew Brooks [40], Alexander J Brucker [41], William H Cade [42,43], Melinda S Cain [24], Peter A Campochiaro [11,44], Chi-Chao Chan [45], Ching-Yu Cheng [34,35,46,47], Emily Y Chew [48], Kimberly A Chin [7], Itay Chowers [49], David G Clayton [50], Radu Cojocaru [40], Yvette P Conley [51], Belinda K Cornes [34], Mark J Daly [15], Baljean Dhillon [30], Albert O Edwards [52], Evangelos Evangelou [53], Jesen Fagerness [54,55], Henry A Ferreyra [56], James S Friedman [40], Asbjorg Geirsdottir [57], Ronnie J George [58], Christian Gieger [59], Neel Gupta [40], Stephanie A Hagstrom [60], Simon P Harding [61], Christos Haritoglou [62], John R Heckenlively [39], Frank G Holz [63], Guy Hughes [56,64], John P A Ioannidis [65,66,67], Tatsuro Ishibashi [68], Peronne Joseph [17,18], Gyungah Jun [4,69,70], Yoichiro Kamatani [71], Nicholas Katsanis [72,73,74], Claudia N Keilhauer [75], Jane C Khan [50,76,77], Ivana K Kim [78,79], Yutaka Kiyohara [80], Barbara E K Klein [81], Ronald Klein [81], Jaclyn L Kovach [82], Igor Kozak [56], Clara J Lee [56,64], Kristine E Lee [81], Peter Lichtner [83], Andrew J Lotery [84], Thomas Meitinger [83,85], Paul Mitchell [86], Saddek Mohand-Saïd [31,32,36,87], Anthony T Moore [14], Denise J Morgan [88], Margaux A Morrison [88], Chelsea E Myers [81], Adam C Naj [42,43], Yusuke Nakamura [89], Yukinori Okada [90], Anton Orlin [91], M Carolina Ortube [92,93], Mohammad I Othman [39], Chris Pappas [88], Kyu Hyung Park [94], Gayle J T Pauer [60], Neal S Peachey [60,95], Olivier Poch [96], Rinki Ratna Priya [40], Robyn Reynolds [7], Andrea J Richardson [24], Raymond Ripp [96], Guenther Rudolph [62], Euijung Ryu [97], José-Alain Sahel [10,31,32,33,36,98,99], Debra A Schaumberg [78,100], Hendrik P N Scholl [44,63], Stephen G Schwartz [82], William K Scott [42,43], Humma Shahid [50,101], Haraldur Sigurdsson [57,102], Giuliana Silvestri [103], Theru A Sivakumaran [104], R Theodore Smith [25,105], Lucia Sobrin [78,79], Eric H Souied [106], Dwight E Stambolian [107], Hreinn Stefansson [8], Gwen M Sturgill-Short [95], Atsushi Takahashi [90], Nirubol Tosakulwong [97], Barbara J Truitt [17,18], Evangelia E Tsironi [108], André G Uitterlinden [19,109], Cornelia M van Duijn [19], Lingam Vijaya [58], Johannes R Vingerling [19,20], Eranga N Vithana [34,35], Andrew R Webster [14], H.-Erich Wichmann [110,111,112,113], Thomas W Winkler [114], Tien Y Wong [24,34,35], Alan F Wright [115], Diana Zelenika [116], Li Zhang [56,64,117], Ling Zhao [56,64], Kang Zhang [56,64,117], Michael L Klein [26], Gregory S Hageman [88,118], G Mark Lathrop [71,116], Kari Stefansson [8,102], Rando Allikmets [25,119,+], Paul N Baird [24,+], Michael B Gorin [92,93,120,+], Jie Jin Wang [24,86,+], Caroline C W Klaver [19,20,+], Johanna M Seddon [7,121,+], Margaret A Pericak-Vance [42,43,+], Sudha K Iyengar [17,18,122,123,124,+], John R W Yates [14,50,+], Anand Swaroop [39,40,+], Bernhard H F Weber [1,+], Michiaki Kubo [13,+], Margaret M DeAngelis [88,+], Thierry Léveillard [10,31,32,+], Unnur Thorsteinsdottir [8,102,+], Jonathan L Haines [5,6,+], Lindsay A Farrer [4,69,70,125,126,+], Iris M Heid [59,114,+], Gonçalo R Abecasis [2,+]

AFFILIATIONS

1Institute of Human Genetics, University of Regensburg, Regensburg, Germany.

2Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, USA.

3Division of Pediatric Pulmonary Medicine, Allergy and Immunology, Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.

4Department of Medicine (Section of Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA.

5Center for Human Genetics Research, Vanderbilt University Medical Center, Nashville, Tennessee, USA.

6Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.

7Ophthalmic Epidemiology and Genetics Service, Tufts Medical Center, Boston, Massachusetts, USA.

8deCODE genetics, Reykjavik, Iceland.

9Department of Molecular Biology and Genetics, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.

10Department of Genetics, Institut de la Vision, UPMC Univ Paris 06, UMR_S 968, Paris, France.

11Department of Neuroscience, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.

12Institute of Genetic Medicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.

13Laboratory for Genotyping Development, Research Group for Genotyping, Center for Genomic Medicine (CGM), RIKEN, Yokohama, Japan.

14Moorfields Eye Hospital and Institute of Ophthalmology, University College London, London, UK.

15Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.

16Stanley Center for Psychiatric Research, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.

17Department of Epidemiology, Case Western Reserve University, Cleveland, Ohio, USA.

18Department of Biostatistics, Case Western Reserve University, Cleveland, Ohio, USA.

19Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands.

20Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.

21Centre for Molecular Epidemiology, National University of Singapore, Singapore, Singapore.

22Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.

23Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.

24Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia.

25Department of Ophthalmology, Columbia University, New York, New York, USA.

26Macular Degeneration Center, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA.

27Department of Bioengineering, University of California San Diego, La Jolla, California, USA.

28Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA.

29Department of Ophthalmology & Visual Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.

30Department of Ophthalmology, University of Edinburgh and Princess Alexandra Eye Pavilion, Edinburgh, UK.

31INSERM, U968, Paris, France.

32CNRS, UMR_7210, Paris, France.

33Institute of Ophthalmology, University College London, London, UK.

34Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore.

35Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.

36Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France.

37School of Biomedicine, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK.

38Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.

39Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, USA.

40Neurobiology Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA.

41Penn Presbyterian Medical Center, Scheie Eye Institute, Philadelphia, Pennsylvania, USA.

42John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, USA.

43Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA.

44Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.

45Immunopathology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA.

46Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore.

47Centre for Quantitative Medicine, Office of Clinical Sciences, Duke-NUS Graduate Medical School, Singapore, Singapore.

48Division of Epidemiology and Clinical Applications, the Clinical Trials Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA.

49Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.

50Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.

51Department of Health Promotion and Development, School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.

52Institute for Molecular Biology, University of Oregon, Eugene, Oregon, USA.

53Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece.

54Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA.

55Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.

56Department of Ophthalmology and Shiley Eye Center, University of California San Diego, La Jolla, California, USA.

57Department of Ophthalmology, National University Hospital, Reykjavik, Iceland.

58Department of Glaucoma, Vision Research Foundation, Sankara Nethralaya, Chennai, India.

59Institute of Genetic Epidemiology, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany.

60Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA.

61Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK.

62Augenklinik, Ludwig-Maximilians-Universität München, München, Germany.

63Department of Ophthalmology, University of Bonn, Bonn, Germany.

64Institute for Genomic Medicine, University of California San Diego, La Jolla, California, USA.

65Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.

66Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California, USA.

67Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, California, USA.

68Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan.

69Department of Ophthalmology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA.

70Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA.

71Fondation Jean Dausset, Centre d’Etude du Polymorphisme Humain (CEPH), Paris, France.

72Center for Human Disease Modeling, Duke University, Durham, North Carolina, USA.

73Department of Cell Biology, Duke University, Durham, North Carolina, USA.

74Department of Pediatrics, Duke University, Durham, North Carolina, USA.

75Department of Ophthalmology, Julius-Maximilians-Universität, Würzburg, Germany.

76Department of Ophthalmology, Royal Perth Hospital, Perth, Australia.

77Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia.

78Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA.

79Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA.

80Department of Environmental Medicine, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan.

81Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.

82Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA.

83Institute of Human Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany.

84Faculty of Medicine, Clinical and Experimental Sciences, University of Southampton, Southampton, UK.

85Institute of Human Genetics, Technische Universität München, München, Germany.

86Centre for Vision Research, Department of Ophthalmology and the Westmead Millennium Institute, University of Sydney, Sydney, Australia.

87Department of Therapeutics, Institut de la Vision, UPMC Univ Paris 06, UMR_S 968, Paris, France.

88Department of Ophthalmology and Visual Sciences, University of Utah, John A. Moran Eye Center, Salt Lake City, Utah, USA.

89Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan.

90Laboratory for Statistical Analysis, Center for Genomic Medicine (CGM), RIKEN, Yokohama, Japan.

91Department of Ophthalmology Weill Cornell Medical College, Weill Cornell Medical College, New York, New York, USA.

92Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.

93Jules Stein Eye Institute, Los Angeles, California, USA.

94Department of Ophthalmology, Seoul National University Bundang Hospital, Kyeounggi, Rep. of Korea.

95Research Service, Louis Stokes Veteran Affairs Medical Center, Cleveland, Ohio, USA.

96Laboratory of Integrative Bioinformatics and Genomics, IGBMC, Illkirch, France.

97Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA.

98Fondation Ophtalmologique Adolphe de Rothschild, Paris, France.

99Académie des Sciences–Institut de France, Paris, France.

100Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA.

101Department of Ophthalmology, Addenbrooke’s Hospital, Cambridge, UK.

102Faculty of Medicine, University of Iceland, Reykjavik, Iceland.

103Centre for Vision and Vascular Science, Queen’s University, Belfast, UK.

104Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.

105Department of Biomedical Engineering, Columbia University, New York, New York, USA.

106Hôpital Intercommunal de Créteil, Hôpital Henri Mondor, Université Paris Est, Créteil, France.

107Department of Ophthalmology and Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.

108Department of Ophthalmology, University of Thessaly School of Medicine, Larissa, Greece.

109Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands.

110Institute of Epidemiology I, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany.

111Institute of Medical Informatics, Ludwig-Maximilians-Universität, and Klinikum Grosshadern, München, Germany.

112Institute of Biometry, Ludwig-Maximilians-Universität, and Klinikum Grosshadern, München, Germany.

113Institute of Epidemiology, Ludwig-Maximilians-Universität, and Klinikum Grosshadern, München, Germany.

114Department of Epidemiology and Preventive Medicine, Regensburg University Medical Center, Regensburg, Germany.

115Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Edinburgh, UK.

116Centre National de Génotypage, CEA - IG, Evry, France.

117Molecular Medicine Research Center and Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China.

118Center for Translational Medicine, University of Utah, John A. Moran Eye Center, Salt Lake City, Utah, USA.

119Department of Pathology & Cell Biology, Columbia University, New York, New York, USA.

120Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.

121Tufts University School of Medicine, Boston, Massachusetts, USA.

122Department of Genetics, Case Western Reserve University, Cleveland, Ohio, USA.

123Department of Ophthalmology, Case Western Reserve University, Cleveland, Ohio, USA.

124Department of Clinical Investigation, Case Western Reserve University, Cleveland, Ohio, USA.

125Department of Neurology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA.

126Department of Epidemiology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA.

*These authors contributed equally to this work.

+These authors share starred senior authorship. They directed the project or one of its major component studies.

AUTHOR CONTRIBUTIONS

AMD Gene Analysis Committee: L.G.F., W.C., M.S., B.L.Y., Y.Y., L.A.F., I.M.H. (co-lead), G.R.A. (co-lead);

AMD Gene Steering Committee: B.H.F.W. (chair, senior executive committee), G.R.A. (senior executive committee), M.M.D. (senior executive committee), J.L.H. (senior executive committee), S.K.I. (senior executive committee), M.A.P. (senior executive committee), R.A., P.N.Ba., C.C.W.K., B.E.K.K., M.L.K., M.K., T.L., J.M.S., U.T., D.E.W., J.R.W.Y., K.Z.;

AMD-EU-JHU Study: D.J.Z., I.A., M.Be., A.C.B., P.A.C., I.C., F.G.H., Y.Ka., N.K., A.J.L., S.M., O.P., R.Ri., J-A.S., H.P.N.S., E.H.S., A.R.W., D.Z., G.M.L., T.L. contributed phenotype, genotypes and analysis for the AMD-EU-JHU study;

BDES Study: R.P.I., B.E.K.K., R.K., K.E.L., C.E.M., T.A.S., B.J.T., S.K.I. contributed phenotype, genotypes and analysis for the BDES study;

Blue Mountains Eye Study: X.S., P.M., T.Y.W., J.J.W. contributed phenotype, genotypes and analysis for the BMES study;

BU/Utah Study: M.S., G.S.H., G.J., I.K.K., D.J.M., M.A.M., C.P., K.H.P., D.A.S., G.S., E.E.T., M.M.D., L.A.F. contributed phenotype, genotypes and analysis for the BU/UTAH study;

CCF/VAMC Study: S.A.H., P.J., G.J.T.P., N.S.P., G.M.S., R.P.I., S.K.I. contributed phenotype, genotypes and analysis for the CCF/VAMC study;

CEI Study: P.J.F., M.L.K. contributed phenotype, genotypes and analysis for the CEI study;

Columbia Study: J.E.M., G.R.B., R.T.S., R.A. contributed phenotype, genotypes and analysis for the Columbia study;

deCode: G.T., H.Si., H.St., K.S., U.T. contributed phenotype, genotypes and analysis for the deCode study;

Japan Age Related Eye Diseases Study: S.A., T.I., Y.Ki., Y.N., Y.O., A.T., M.K. contributed phenotype, genotypes and analysis for the JAREDS study;

Melbourne Study: R.H.G., M.R.N.C., A.J.R., P.N.Ba. contributed phenotype, genotypes and analysis for the Melbourne study;

Miami/Vanderbilt Study: B.L.Y., A.A., W.H.C., J.L.K., A.C.N., S.G.S., W.K.S., M.A.P., J.L.H. contributed phenotype, genotypes and analysis for the Miami/Vanderbilt study;

MMAP/NEI Study: W.C., K.E.B., M.Br., A.J.B., C-C.C., E.Y.C., R.C., A.O.E., J.S.F., N.G., J.R.H., A.O., M.I.O., R.R.P., E.R., D.E.S., N.T., A.S., G.R.A. contributed phenotype, genotypes and analysis for the MMAP/NEI study;

Rotterdam Study: G.H.S.B., A.G.U., C.M.v.D., J.R.V., C.C.W.K. contributed phenotype, genotypes and analysis for the Rotterdam study;

SAGE Study: T.A., C-Y.C., B.K.C., E.N.V. contributed phenotype, genotypes and analysis for the SAGe study;

Southern German AMD Study: L.G.F., C.G., C.H., C.N.K., P.L., T.M., G.R., H.-E.W., T.W.W., B.H.F.W., I.M.H. contributed phenotype, genotypes and analysis for the Southern German AMD study;

Tufts/Massachussets General Hospital Study: Y.Y., S.R., K.A.C., M.J.D., E.E., J.F., J.P.A.I., R.Re., L.S., J.M.S. contributed phenotype, genotypes and analysis for the Tufts/MGH study;

U.K. Cambridge/Edinburgh Study: V.C., A.M.A., P.N.Bi., D.G.C., B.D., S.P.H., J.C.K., A.T.M., H.Sh., A.F.W., J.R.W.Y. contributed phenotype, genotypes and analysis for the UK Cambridge/Edinburgh study;

University of Pittsburgh/UCLA Study: D.E.W., Y.P.C., M.C.O., M.B.G. contributed phenotype, genotypes and analysis for the Univ. of Pittsburgh/UCLA study;

UCSD Study: G.Ha., H.F., G.Hu., I.K., C.J.L., L.Zhang, L.Zhao, K.Z. contributed phenotype, genotypes and analysis for the USCD study;

VRF Study: R.J.G., L.V., R.P.I., S.K.I. contributed phenotype, genotypes and analysis for the VRF study;

Gene Expression and RNA-Sequencing Data: These data were contributed and analyzed by M.Br., J.S.F., N.G., R.R.P and A.S.

URLs

METAL, http://www.sph.umich.edu/csg/abecasis/Metal/; R, http://www.R-project.org/; gee, http://CRAN.R-project.org/package=gee; Single Nucleotide Polymorphism Spectral Decomposition, http://gump.qimr.edu.au/general/daleN/SNPSpDlite/; Pre-specified R-scripts, http://www.epi-regensburg.de/wp/genepi-downloads; The 1000 Genomes Project, http://www.1000genomes.org/; The HapMap Project, http://www.hapmap.org/genotypes/; PolyPhen-2, http://genetics.bwh.harvard.edu/pph2/; Ingenuity® Systems, http://www.ingenuity.com; NHGRI GWAS catalog, http://www.genome.gov/gwastudies/, INRICH, http://atgu.mgh.harvard.edu/inrich; Full Result Set, http://www.sph.umich.edu/csg/abecasis/public/amdgene2012/

CONFLICT OF INTEREST STATEMENT

A.A., G.R.A., K.E.B., V.C., Y.P.C., M.J.D., A.O.E., L.G.F., M.B.G., J.L.H., A.T.M., D.A.S., W.K.S., J.M.S., A.S., B.H.F.W., D.E.W., and J.R.W.Y. are co-inventors or beneficiaries of patents related to genetic discoveries in AMD. J.L.H. is a shareholder in ArcticDX. S.G.S. is a consultant for Alimera, Bausch + Lomb, and Eyetech; receives royalties from IC Labs. U.T., K.S., G.T, and H.St. are affiliated and/or employed by deCODE Genetics and own stock and/or stock options in the company. P.M. is on advisory boards for Allergan, Bayer, Novartis, Pfizer and Solvay and has received travel, honorarium and research support from these companies; he has no stocks, equity, contract of employment or named position on company board.

Acknowledgments

We are indebted to all the participants who volunteered their time, DNA and information to make this research study possible. We are also in great debt to the clinicians, nurses and research staff who participated in patient recruitment and phenotyping. We thank Dr. Hemin Chin for constant support and encouragement, which helped us bring this project to completion. We thank Drs. Sheldon Miller and Jennifer Barb for access to RPE expression data and the MIGEN study group for use of their genotype data. We thank C. Pappas, N. Miller, J. Hageman, W. Hubbard, L. Lucci, A. Vitale, P. Bernstein and N. Amin for technical and clinical assistance. We thank E Rochtchina, AC Viswanathan, J Xie, M Inouye, EG Holliday, J Attia, RJ Scott for contributions to the Blue Mountains Eye Study GWAS. We thank members of the Genetic Factors in AMD Study Group, the Scottish Macula Society Study Group, the Wellcome Trust Clinical Research facility at Southampton General Hospital. We thank Tunde Peto and colleagues at the Reading Centre, Moorfields Eye Hospital, London, and C. Brussee and A. Hooghart for help in patient recruitment and phenotyping. Full details of funding sources can be found is in the Supplementary Information.

References

  • 1. SwaroopAChewEYRickmanCBAbecasisGRUnravelling a Late-Onset Multifactorial Disease: From Genetic Susceptibility to Disease Mechanisms for Age-related Macular DegenerationAnnu Rev Genomics Hum Genet1019432009[PubMed][Google Scholar]
  • 2. SeddonJMCoteJPageWFAggenSHNealeMCThe US twin study of age-related macular degeneration: relative roles of genetic and environmental influencesArchives of Ophthalmology12332172005[PubMed][Google Scholar]
  • 3. FriedmanDSPrevalence of age-related macular degeneration in the United StatesArchives of Ophthalmology122564722004[PubMed][Google Scholar]
  • 4. EdwardsAOComplement factor H polymorphism and age-related macular degenerationScience30842142005[PubMed][Google Scholar]
  • 5. HainesJLComplement factor H variant increases the risk of age-related macular degenerationScience308419212005[PubMed][Google Scholar]
  • 6. KleinRJComplement factor H polymorphism in age-related macular degenerationScience30838592005[PubMed][Google Scholar]
  • 7. YatesJRComplement C3 variant and the risk of age-related macular degenerationN Engl J Med357553612007[PubMed][Google Scholar]
  • 8. GoldBVariation in factor B (BF) and complement component 2 (C2) genes is associated with age-related macular degenerationNat Genet38458622006[PubMed][Google Scholar]
  • 9. FagernessJAVariation near complement factor I is associated with risk of advanced AMDEur J Hum Genet1710042009[PubMed][Google Scholar]
  • 10. HagemanGSA common haplotype in the complement regulatory gene factor H (HF1/CFH) predisposes individuals to age-related macular degenerationProc Natl Acad Sci U S A1027227322005[PubMed][Google Scholar]
  • 11. MallerJBVariation in complement factor 3 is associated with risk of age-related macular degenerationNat Genet39120012007[PubMed][Google Scholar]
  • 12. RiveraAHypothetical LOC387715 is a second major susceptibility gene for age-related macular degeneration, contributing independently of complement factor H to disease riskHum Mol Genet143227362005[PubMed][Google Scholar]
  • 13. JakobsdottirJSusceptibility genes for age-related maculopathy on chromosome 10q26Am J Hum Genet773894072005[PubMed][Google Scholar]
  • 14. KlaverCCGenetic association of apolipoprotein E with age-related macular degenerationAmerican Journal of Human Genetics6320061998[PubMed][Google Scholar]
  • 15. SouiedEHThe epsilon4 allele of the apolipoprotein E gene as a potential protective factor for exudative age-related macular degenerationAmerican Journal of Ophthalmology12535391998[PubMed][Google Scholar]
  • 16. McKayGJEvidence of association of APOE with age-related macular degeneration: a pooled analysis of 15 studiesHuman Mutation321407162011[PubMed][Google Scholar]
  • 17. ChenWGenetic variants near TIMP3 and high-density lipoprotein-associated loci influence susceptibility to age-related macular degenerationProceedings of the National Academy of Sciences of the United States of America107740162010[PubMed][Google Scholar]
  • 18. NealeBMGenome-wide association study of advanced age-related macular degeneration identifies a role of the hepatic lipase gene (LIPC)Proceedings of the National Academy of Sciences of the United States of America10773954002010[PubMed][Google Scholar]
  • 19. YuYCommon variants near FRK/COL10A1 and VEGFA are associated with advanced age-related macular degenerationHuman Molecular Genetics2036997092011[PubMed][Google Scholar]
  • 20. ArakawaSGenome-wide association study identifies two susceptibility loci for exudative age-related macular degeneration in the Japanese populationNature Genetics43100142011[PubMed][Google Scholar]
  • 21. McCarthyMIGenome-wide association studies for complex traits: consensus, uncertainty and challengesNat Rev Genet9356692008[PubMed][Google Scholar]
  • 22. LiYWillerCJSannaSAbecasisGRGenotype ImputationAnnu Rev Genomics Hum Genet103874062009[PubMed][Google Scholar]
  • 23. MarchiniJHowieBMyersSMcVeanGDonnellyPA new multipoint method for genome-wide association studies by imputation of genotypesNature Genetics39906132007[PubMed][Google Scholar]
  • 24. BrowningBLBrowningSRA unified approach to genotype imputation and haplotype-phase inference for large data sets of trios and unrelated individualsAmerican Journal of Human Genetics84210232009[PubMed][Google Scholar]
  • 25. LiYWillerCJDingJScheetPAbecasisGRMaCH: using sequence and genotype data to estimate haplotypes and unobserved genotypesGenetic Epidemiology34816342010[PubMed][Google Scholar]
  • 26. WillerCJLiYAbecasisGRMETAL: fast and efficient meta-analysis of genomewide association scansBioinformatics26219012010[PubMed][Google Scholar]
  • 27. SkolADScottLJAbecasisGRBoehnkeMJoint analysis is more efficient than replication-based analysis for two-stage genome-wide association studiesNat Genet38209132006[PubMed][Google Scholar]
  • 28. HigginsJPThompsonSGDeeksJJAltmanDGMeasuring inconsistency in meta-analysesBMJ327557602003[PubMed][Google Scholar]
  • 29. SobrinLARMS2/HTRA1 locus can confer differential susceptibility to the advanced subtypes of age-related macular degenerationAmerican Journal of Ophthalmology15134552e32011[PubMed][Google Scholar]
  • 30. SeddonJMAssociation of CFH Y402H and LOC387715 A69S with progression of age-related macular degenerationJAMA29717938002007[PubMed][Google Scholar]
  • 31. LiMCFH haplotypes without the Y402H coding variant show strong association with susceptibility to age-related macular degenerationNature Genetics381049542006[PubMed][Google Scholar]
  • 32. MallerJCommon variation in three genes, including a noncoding variant in CFH, strongly influences risk of age-related macular degenerationNature Genetics38105592006[PubMed][Google Scholar]
  • 33. NejentsevSWalkerNRichesDEgholmMToddJARare variants of IFIH1, a gene implicated in antiviral responses, protect against type 1 diabetesScience32438792009[PubMed][Google Scholar]
  • 34. RaychaudhuriSA rare penetrant mutation in CFH confers high risk of age-related macular degenerationNature Genetics43123262011[PubMed][Google Scholar]
  • 35. PruimRJLocusZoom: regional visualization of genome-wide association scan resultsBioinformatics26233672010[PubMed][Google Scholar]
  • 36. AdzhubeiIAA method and server for predicting damaging missense mutationsNat Methods724892010[PubMed][Google Scholar]
  • 37. SivakumaranTAA 32 kb Critical Region Excluding Y402H in CFH Mediates Risk for Age-Related Macular DegenerationPLoS ONE6e255982011[PubMed][Google Scholar]
  • 38. The Wellcome Trust Case Control ConsortiumGenome-wide association study of CNVs in 16, 000 cases of eight common diseases and 3, 000 shared controlsNature464713202010[PubMed][Google Scholar]
  • 39. The 1000 Genomes ProjectA map of human genome variation from population scale sequencingNature4672010[Google Scholar]
  • 40. FritscheLGAge-related macular degeneration is associated with an unstable ARMS2 (LOC387715) mRNANat Genet4089262008[PubMed][Google Scholar]
  • 41. DewanAHTRA1 promoter polymorphism in wet age-related macular degenerationScience314989922006[PubMed][Google Scholar]
  • 42. HughesAEA common CFH haplotype, with deletion of CFHR1 and CFHR3, is associated with lower risk of age-related macular degenerationNat Genet38117372006[PubMed][Google Scholar]
  • 43. FritscheLGAn imbalance of human complement regulatory proteins CFHR1, CFHR3 and factor H influences risk for age-related macular degeneration (AMD)Human Molecular Genetics1946947042010[PubMed][Google Scholar]
  • 44. BrooksMJRajasimhaHKRogerJESwaroopANext-generation sequencing facilitates quantitative analysis of wild-type and Nrl(−/−) retinal transcriptomesMolecular Vision173034542011[PubMed][Google Scholar]
  • 45. StrunnikovaNVTranscriptome analysis and molecular signature of human retinal pigment epitheliumHuman Molecular Genetics192468862010[PubMed][Google Scholar]
  • 46. HindorffLAPotential etiologic and functional implications of genome-wide association loci for human diseases and traitsProceedings of the National Academy of Sciences of the United States of America106936272009[PubMed][Google Scholar]
  • 47. SubramanianAGene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profilesProceedings of the National Academy of Sciences of the United States of America10215545502005[PubMed][Google Scholar]
  • 48. ManolioTAFinding the missing heritability of complex diseasesNature461747532009[PubMed][Google Scholar]
  • 49. SoHCGuiAHChernySSShamPCEvaluating the heritability explained by known susceptibility variants: a survey of ten complex diseasesGenetic Epidemiology3531072011[PubMed][Google Scholar]
  • 50. SeddonJMReynoldsRYuYDalyMJRosnerBRisk models for progression to advanced age-related macular degeneration using demographic, environmental, genetic, and ocular factorsOphthalmology1182203112011[PubMed][Google Scholar]
  • 51. DevlinBRoederKGenomic control for association studiesBiometrics5599710041999[PubMed][Google Scholar]
  • 52. WallaceCThe imprinted DLK1-MEG3 gene region on chromosome 14q32.2 alters susceptibility to type 1 diabetesNature Genetics4268712010[PubMed][Google Scholar]
  • 53. HuangLGenotype-imputation accuracy across worldwide human populationsAmerican Journal of Human Genetics84235502009[PubMed][Google Scholar]
  • 54. PurcellSPLINK: a tool set for whole-genome association and population-based linkage analysesAm J Hum Genet81559752007[PubMed][Google Scholar]
  • 55. AulchenkoYSStruchalinMVvan DuijnCMProbABEL package for genome-wide association analysis of imputed dataBMC Bioinformatics111342010[PubMed][Google Scholar]
  • 56. ZegerSLLiangKYLongitudinal data analysis for discrete and continuous outcomesBiometrics42121301986[PubMed][Google Scholar]
  • 57. CareyVJPorted to R by Thomas Lumley and Brian Ripley. Note that maintainers are not available to give advice on using a package they did not author. gee: Generalized Estimation Equation solverR package version 4.13–182012[Google Scholar]
  • 58. R Core TeamR: A language and environment for statistical computingR Foundation for Statistical ComputingVienna, Austria3-900051-07-02012
  • 59. The International HapMap Consortium et alIntegrating common and rare genetic variation in diverse human populationsNature4675282010[PubMed][Google Scholar]
  • 60. NyholtDRA simple correction for multiple testing for single-nucleotide polymorphisms in linkage disequilibrium with each otherAm J Hum Genet7476592004[PubMed][Google Scholar]
  • 61. The 1000 Genomes Project Consortium et alAn integrated map of genetic variation from 1, 092 human genomesNature49156652012[PubMed][Google Scholar]
  • 62. WangKLiMHakonarsonHANNOVAR: functional annotation of genetic variants from high-throughput sequencing dataNucleic Acids Res38e1642010[PubMed][Google Scholar]
  • 63. LeePHO’DushlaineCThomasBPurcellSMINRICH: interval-based enrichment analysis for genome-wide association studiesBioinformatics28179792012[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.