Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Oct 1.
Published in final edited form as: Nat Genet. 2011 Sep 18;43(10):969–976. doi: 10.1038/ng.940

Genome-wide association study identifies five new schizophrenia loci

The Schizophrenia Psychiatric Genome-Wide Association Study (GWAS) Consortium1
PMCID: PMC3303194  NIHMSID: NIHMS360727  PMID: 21926974

Abstract

We examined the role of common genetic variation in schizophrenia in a genome-wide association study of substantial size: a stage 1 discovery sample of 21,856 individuals of European ancestry and a stage 2 replication sample of 29,839 independent subjects. The combined stage 1 and 2 analysis yielded genome-wide significant associations with schizophrenia for seven loci, five of which are new (1p21.3, 2q32.3, 8p23.2, 8q21.3 and 10q24.32-q24.33) and two of which have been previously implicated (6p21.32-p22.1 and 18q21.2). The strongest new finding (P = 1.6 × 10−11) was with rs1625579 within an intron of a putative primary transcript for MIR137 (microRNA 137), a known regulator of neuronal development. Four other schizophrenia loci achieving genome-wide significance contain predicted targets of MIR137, suggesting MIR137-mediated dysregulation as a previously unknown etiologic mechanism in schizophrenia. In a joint analysis with a bipolar disorder sample (16,374 affected individuals and 14,044 controls), three loci reached genome-wide significance: CACNA1C (rs4765905, P = 7.0 × 10−9), ANK3 (rs10994359, P = 2.5 × 10−8) and the ITIH3-ITIH4 region (rs2239547, P = 7.8 × 10−9).

In stage 1, we conducted a mega-analysis combining genome-wide assocation study (GWAS) data from 17 separate studies (with a total of 9,394 cases and 12,462 controls; Table 1 and Supplementary Tables 1,2). We imputed allelic dosages for 1,252,901 autosomal SNPs (Table 1, Supplementary Table 3 and Supplementary Note) using HapMap3 as the reference panel1. We tested for association using logistic regression of imputed dosages with sample identifiers and three principal components as covariates to minimize inflation in significance testing caused by population stratification. The quantile-quantile plot (Supplementary Fig. 1) deviated from the null distribution with a population stratification inflation factor of λ = 1.23. However, λ1000, a metric that standardizes the degree of inflation by sample size, was only 1.02, similar to that observed in other GWAS meta-analyses2,3. This deviation persisted despite comprehensive quality control and inclusion of up to 20 principal components (Supplementary Fig. 1). Thus, we interpret this deviation as indicative of a large number of weakly associated SNPs consistent with polygenic inheritance4. We also examined 298 ancestry-informative markers (AIMs) that reflect European-ancestry population substructure5. Unadjusted analyses showed greater inflation in the test statistics than we saw for all markers (AIMs λ = 2.26 compared to all markers λ = 1.56). After inclusion of principal components, the distributions of the test statistics did not differ between AIMs (λ = 1.18) and all markers (λ = 1.23), a result inconsistent with population stratification explaining the residual deviation seen in Supplementary Figure 1. Moreover, the results of a meta-analysis using summary results generated using study specific principal components (Supplementary Note) were highly correlated with those from the mega-analysis (Pearson correlation = 0.94, with a similar λ = 1.20; Supplementary Fig. 2). Of the ten SNPs in Table 2, four increased and six decreased in significance, suggesting that the most extreme values did not result from systematic inflation artifacts. Therefore, our primary analysis used unadjusted P values (nevertheless, see Table 2 for stage 1 P values adjusted for λ (ref. 6).

Table 1.

Study design and samples

Collection Country Platform Cases included by sex
Controls included by sex
Male Female Unknown Total Male Female Unknown Total
Cardiff UK UK Affymetrix 500K 320 152 0 472 1,442 1,492 0 2,934
CATIE United States Affymetrix 500K; Perlegen 164K 308 94 0 402 161 46 0 207
ISC-Aberdeen UK Affymetrix 5.0 536 184 0 720 447 251 0 698
ISC-Cardiff Bulgaria Affymetrix 6.0 270 257 0 527 291 318 0 609
ISC-Dublin Ireland Affymetrix 6.0 188 82 0 270 258 602 0 860
ISC-Edinburgh UK Affymetrix 6.0 267 101 0 368 146 138 0 284
ISC-London UK Affymetrix 5.0; Affymetrix 500K 369 149 0 518 207 284 0 491
ISC-Portugal Portugal Affymetrix 5.0 213 133 0 346 80 135 0 215
ISC-SW1 Sweden Affymetrix 5.0 93 75 0 168 82 85 0 167
ISC-SW2 Sweden Affymetrix 6.0 231 159 0 390 116 113 0 229
MGS United States, Australia Affymetrix 6.0 1,863 816 0 2,679 1,140 1,344 0 2,484
SGENE-Bonn Germany Illumina 550K 238 236 0 474 664 640 0 1,304
SGENE-Copenhagen Denmark Illumina Human 610-Quad 280 202 0 482 268 189 0 457
SGENE-Munich Germany Illumina 300K 279 155 0 434 167 184 0 351
SGENE-TOP3 Norway Affymetrix 6.0 132 116 0 248 176 175 0 351
SGENE-UCLA The Netherlands Illumina 550K 529 175 0 704 310 321 631
Zucker Hillside United States Affymetrix 500K 128 64 0 192 92 98 0 190
Grand totals for the GWAs 6,244 3,150 0 9,394 6,047 6,415 12,462

Multicenter Pedigree Europe, United States, Australia Illumina Human 610-Quad n.a. n.a. 0 583 0 0 0 0
SGENE-Aarhus Denmark Illumina Human 610-Quad 477 399 0 876 477 397 0 874
SGENE-Aarhus Denmark Centaurus 114 102 1 217 176 317 0 493
SGENE-Belgium Belgium Centaurus; Illumina 370K 326 184 0 510 149 192 0 341
SGENE-Copenhagen Denmark Centaurus 264 198 0 462 499 375 0 874
SGENE-Iceland Iceland Illumina 300K 346 185 0 531 5,802 5,813 0 11,615
SGENE-England UK Illumina 300K 71 22 0 93 48 40 0 88
SGENE-Helsinki Finland Illumina 300K 112 70 0 59 122 75 0 147
SGENE-Kuusamo Finland Illumina 300K 123 50
SGENE-Hungary Hungary Centaurus 105 136 0 241 89 125 0 214
SGENE-Italy Italy Illumina 300K 48 36 0 84 50 39 0 89
SGENE-Munich Germany Illumina 300K 280 186 0 163 887 912 0 185
SGENE-Munich Germany Centaurus 303 1,614
SGENE-Russia Russia Centaurus 132 343 0 475 178 290 0 468
SGENE-Sweden Sweden Centaurus 158 94 0 252 178 109 0 287
SW3 Sweden Affymetrix 6.0 327 212 0 539 457 448 0 905
SW4 Sweden Affymetrix 6.0 656 407 0 1,063 605 568 0 1,173
UQ and ASRB Australia SequenomMassArray 347 190 21 558 487 455 15 957
ISGC and WTCCC2 Ireland Affymetrix 6.0 968 342 0 1,310 245 778 0 1,023
Grand totals for the replication follow up 4,731 3,106 22 8,442 10,449 10,933 15 21,397

Stage 1 describes the 17 samples that provided full GWAS genotyping data, and stage 2 describes the 19 studies that provided results for the top SNPs identified in the combined analysis of stage 1 studies. Stage 2 replication SGENE-Belgium had four cases missing sex information. Stage 2 replication SGENE-Aarhus (focused genotyping sample) had one case missing sex information. Stage 2 replication-University of Queensland had 21 cases and 15 controls missing sex information. Sex information for the two stage 2 replication SGENE-Munich samples are combined. Sex information for the two stage 2 replication SGENE-Finnish (Helsinki and Kuusamo) samples are combined to enable that these two samples are located adjacent to each other in the table (rather than alphabetically). Multicenter Pedigree was a family sample, and so case sex counts are not applicable (n.a). SGENE, Schizophrenia Genetics Consortium; ISC, International Schizophrenia Consortium; TOP3, Thematic Organized Psychoses Research 3; UCLA, University of California at Los Angeles; SW1, Sweden 1; SW2, Sweden 2; WTCCC, Wellcome Trust case Control Consortium; for the Multicenter Pedigree study, the number of cases indicates the number of families; CATIE, Clinical Antipsychotic Trials of Intervention Effectiveness; MGS, Molecular Genetics of Schizophrenia; UQ, University of Queensland; ASRB, Australian Schizophrenia Research Bank; ISGC, Irish Schizophrenia Genomics Consortium.

Table 2.

Top genome-wide association results for schizophrenia

SNP Chr. Mb Alleles Frequency P (GC-adjusted P ) OR (95% CI) Consistency of direction Gene Distance (kb)
rs1625579 1p21.3a 98.3 TG 0.80 5.72 × 10−7 (6.52 × 10−6) 1.14 (1.08–1.19) +−++++−+ MIR137 Intragenic
2.65 × 10−6 (n.a.) 1.11 (1.07–1.16)
1.59 × 10−11 (6.87 × 10−10) 1.12 (1.09–1.16)
rs17662626 2q32.3a 193.7 AG 0.91 3.09 × 10−6 (2.60 × 10−5) 1.22 (1.13–1.30) + −+ +++ PCGEM1 343
1.70 × 10−3 (n.a.) 1.16 (1.06–1.27)
4.65 × 10−8 (1.25 × 10−6) 1.20 (1.13–1.26)
rs2021722 6p21.3-p22.1 30.3 CT 0.78 4.30 × 10−11 (2.76 × 10−9) 1.18 (1.13–1.23) + ++ −++ TRIM26 Intragenic
1.55 × 10−3 (n.a.) 1.10 (1.03–1.17)
2.18 × 10−12 (2.88 × 10−10) 1.15 (1.11–1.19)
rs10503253 8p23.2a 4.2 AC 0.19 3.84 × 10−7 (4.71 × 10−6) 1.14 (1.09–1.19) + ++ +−+ CSMD1 Intragenic
7.60 × 10−3 (n.a.) 1.08 (1.01–1.14)
4.14 × 10−8 (8.98 × 10−7) 1.11 (1.07–1.15)
rs7004633 8q21.3a 89.8 GA 0.18 1.45 × 10−8 (3.22 × 10−7) 1.16 (1.11–1.21) ++−+++−+ MMP16 421
0.011 (n.a.) 1.05 (1.01–1.10)
2.75 × 10−8 (7.03 × 10−7) 1.10 (1.07–1.14)
rs7914558 10q24.32a 104.8 GA 0.59 1.58 × 10−7 (2.27 × 10−6) 1.11 (1.07–1.15) + ++ +++ CNNM2 Intragenic
1.07 × 10−3 (n.a.) 1.08 (1.03–1.13)
1.82 × 10−9 (3.11 × 10−8) 1.10 (1.07–1.13)
rs11191580 10q24.33a 104.9 TC 0.91 2.23 × 10−8 (4.58 × 10−7) 1.22 (1.15–1.29) ++++++++ NT5C2 Intragenic
5.09 × 10−3 (n.a.) 1.09 (1.02–1.16)
1.11 × 10−8 (3.72 × 10−7) 1.15 (1.10–1.20)
rs548181 11q24.2 125.0 GA 0.88 2.91 × 10−8 (5.69 × 10−7) 1.20 (1.13–1.26) +++++−+ STT3A 1
0.068 (n.a.) 1.04 (0.98–1.11)
8.87 × 10−7 (1.74 × 10−5) 1.11 (1.07–1.16)
rs12966547 18q21.2 50.9 GA 0.58 1.00 × 10−6 (1.03 × 10−5) 1.10 (1.06–1.14) +−++++++ CCDC68 126
2.29 × 10−5 (n.a.) 1.08 (1.04–1.12)
2.60 × 10−10 (5.99 × 10−9) 1.09 (1.06–1.12)
rs17512836 18q21.2 51.3 CT 0.02 2.35 × 10−8 (4.78 × 10−7) 1.40 (1.28–1.52) −+++++++ TCF4 Intragenic
0.085 (n.a.) 1.08 (0.96–1.20)
1.05 × 10−6 (2.86 × 10−5) 1.23 (1.14–1.31)

The SNPs listed are those with a stage 1 P < 5 × 10−8 and/or a combined stage 1 and 2 P < 5 × 10−8. These ten independent (r2 < 0.2) SNPs represent eight physically distinct genomic loci, as there are two SNPs listed for two loci (10q24.32-q24.33 and 18q21.2). For the MHC region, only one SNP is listed for clarity. The eight susceptibility loci represent three previously reported and five new loci (Supplementary Table 7). Stage 1 is the discovery GWAS mega-analysis. Stage 2 is the replication sample (single-tailed meta-analysis P values are weighted by 1/s.e.), and because the P values are single tailed, some 95% confidence intervals contain 1 (if 0.10 < P < 0.05). Combined values include stages 1 and 2 (two-tailed meta-analysis P values are weighted by 1/s.e.). For each SNP, P values and odds ratios are listed for stage 1 (top), stage 2 (middle) and combined stage 1 and 2 analysis (bottom) with the genomic control (GC)-adjusted values bracketed (n.a., not applicable for stage 2). Alleles are listed with the stage 1 risk allele first; the frequency (in stage 1 controls) and odds ratio (OR) refer to the stage 1 risk allele. Bolded P values indicate P < 5 × 10−8, except for in the stage 2 data, where bolded values indicate P < 0.05. The directions of association in eight replication samples are represented by + if the associations are in the same direction, − if they are in opposite directions and a blank space if the data are not available. Mb is the base position based on hg18. Cytogenetic bands are listed for each SNP, though because only one of multiple MHC SNPs are listed, a band range is given in that instance. The nearest gene (or microRNA) is listed, with the distance (kb) from the gene (or if the SNP is intragenic) noted. None of these SNPs showed a significant test for hetereogeneity among the samples. Chr., chromosome.

a

New finding.

In stage 1 (Table 2, Supplementary Table 4 and Supplementary Figs. 3 and 4), 136 associations reached genome-wide significance (P < 5 × 10−8)7. The majority of these associations (N = 129) mapped to 5.5 Mb in the extended major histocompatibility complex (MHC, 6p21.32-p22.1), a region of high linkage disequilibrium (LD) previously implicated in schizophrenia in a subset of the samples used here4,8,9. The other stage 1 regions included new regions (10q24.33 and 8q21.3) and previously reported regions (18q21.2 at TCF4 (encoding transcription factor 4) and 11q24.2 (ref. 8)). The signal at 11q24.2 is ~0.85 Mb from NRGN (encoding neurogranin) and is uncorrelated with the previously associated variant near this gene8.

In Table 2 and Supplementary Table 4, we denote regions of association by the most significant marker. Associated SNPs with r2 ≥ 0.2 in HapMap3 (CEU+TSI populations) were not considered independent. However, we noticed instances where multiple SNPs within 250 kb of each other yielded evidence for association (P < 10−5) despite weak LD (r2 < 0.2) between them. For regions with P < 10−6, we performed a conditional analysis using as covariates the dosages of the strongest associated SNP, principal components 1–4 and 6 and study indicator. We observed multiple statistically independent signals at the MHC. Although a number of SNPs within the MHC were potentially independent per HapMap r2 values, only rs9272105 withstood formal conditional analysis, showing P = 1.8 × 10−6 conditional on association to the best SNP, rs2021722 (stage 1 P = 4.3 × 10−11, inter-SNP distance = 2.4 Mb, r2 = 0.01 in HapMap). Excluding the MHC region, we identified six regions with at least one SNP associated at P < 10−5 and a second SNP with a conditionally independent P < 10−3 (Supplementary Table 5). We performed 100 simulations after permuting case-control status randomly within each study. In contrast to the six regions in the real dataset, we never observed more than a single region with co-localized statistically independent signals in any simulated genome-wide scan, indicating our observation is highly unlikely to have occurred by chance.

Noteworthy co-localizing independent signals occurred at three regions (Supplementary Table 5): one region with a genome-wide significant association at 10q24.32-q24.33 (Table 2), a second region that nearly met this threshold at MAD1L1 (encoding mitotic arrest deficient-like 1; rs10226475, P = 5.06 × 10−8; Supplementary Table 4) and a third region at CACNA1C (encoding calcium channel, voltage-dependent, L type, α 1C subunit), the latter of which has previously been associated with bipolar disorder10 and other psychiatric phenotypes including schizophrenia11. The conditionally independent signal at CACNA1C was more significant than any observation made in 100 permutations of the entire experiment (both conditional P < 10−5) and supports CACNA1C in schizophrenia after genome-wide correction (P < 0.01), even without considering these prior reports.

In stage 2, we evaluated in 29,839 independent subjects (8,442 cases and 21,397 controls) the most significant SNPs (N = 81) in each LD region where at least one SNP had surpassed P < 2 × 10−5 (Supplementary Table 6) in the mega-analysis. Of 22 SNPs from the MHC, 5 surpassed the genome-wide significant threshold in stages 1 and 2 combined (minimum P = 2.2 × 10−12 at rs2021722; Supplementary Table 6). Excluding the MHC region, a sign test for consistency between stages 1 and 2 was highly significant (P < 10−6), with the same direction of effect as observed stage 1 also being observed in stage 2 for 49 of 59 SNPs. A Fisher’s combined test revealed the distribution of stage 2 P values was unlikely to have occurred by chance (P < 10−15). We also performed a transmission analysis using the family based Multicenter Pedigree replication sample in conjunction with a GWAS of 622 parent-offspring schizophrenia trios from Bulgaria12, and the stage 1 associated allele was over-transmitted to cases for 44 of the 59 SNPs (one-sided P = 1.0 × 10−4). Thus, the stage 2 replication results are highly consistent with the stage 1 discovery results.

In the combined dataset (stages 1 and 2), five new (1p21.3, 2q32.3, 8p23.2, 8q21.3 and 10q24.32-q24.33) and two previously reported (6p21.32-p22.1 and 18q21.2) loci met genome-wide significance (Figs. 1,2, Table 2, Supplementary Tables 6,7 and Supplementary Fig. 4). After adjusting for λ (ref. 6), four loci (1p21.3, 6p21.32-p22.1, 10q24.32-q24.33 and 18q21.2) remained significant at P ≤ 5 × 10−8. For the primary analyses (unadjusted for λ), the strongest new association was at 1p21.3 (rs1625579; P = 1.6 × 10−11), which is over 100 kb from any RefSeq protein-coding gene but is within intron 3 of AK094607, which contains the primary transcript for MIR137 (ref. 13). The next best locus, 10q24.32 (Supplementary Table 5 and Supplementary Fig. 5), has independent associations 130 kb apart at rs7914558 (P = 1.8 × 10−9) and rs11191580 (P = 1.1 × 10−8), implicating a 0.5-Mb region containing multiple genes (Supplementary Fig. 5). The third best locus, rs7004633 (P = 2.8 × 10−8) on 8q21.3, is 400 kb from the nearest gene (MMP16, encoding matrix metallopeptidase 16). The fourth best locus, rs10503253 (P = 4.4 × 10−8) at 8p23.2, is in an intron of CSMD1 (encoding CUB and Sushi multiple domains 1). Finally, rs17662626 (P = 4.7 × 10−8) at 2q32.3 is intergenic, mapping 300 kb from a non-coding RNA, PCGEM1 (prostate-specific transcript 1)14.

Figure 1.

Figure 1

Manhattan plot for stages 1 and 2. Standard −log10 P plot of the study results. For the stage 1 results, 16 regions with one or more SNP achieving P < 10−6 are highlighted in color and labeled with the name of the nearest gene. SNPs selected for stage 2 replication are highlighted, with the resulting combined P value after replication (that is, after incorporation of stage 2 results) indicated by the large diamonds. Blue highlighting indicates SNPs that were less significantly associated after replication, and pink highlighting indicates SNPs that were more significantly associated after replication.

Figure 2.

Figure 2

Regional association plots for five new schizophrenia loci. Regional P value plots for each of the five new schizophrenia loci: 1p21.3, 2q32.3, 8p23.2, 8q21.3 and 10q24.32-q24.33. Each plot shows the most associated SNP (key SNP) and its genomic region from the first column of Table 2: stage 1 scan results for each SNP ± 200 kb to the key SNP are shown. On the x axis is the genomic position, and on the y axis is −log10 P. Larger SNP symbols indicate higher LD (based on HapMap 3 data) to the key SNP than smaller SNP symbols. Color coding (from red to blue) denotes LD information; see also the legend within the plot.

MIR137 has been implicated in regulating adult neurogenesis15,16 and neuronal maturation17, mechanisms through which variation at this locus could contribute to brain development abnormalities in schizophrenia. Of relevance, two independent schizophrenia imaging studies found MIR137 to be one of three microRNAs with targets significantly enriched for association18. In stage 1, SNPs in or near 301 high-confidence predicted MIR137 targets (with a TargetScan19 probability of conserved targeting ≥0.9) were enriched for association compared with genes matched for size and marker density: 17 predicted MIR137 targets (Supplementary Table 8) had at least one SNP with P < 10−4, which is more than twice as many as the control gene sets (P < 0.01). Excluding the MHC and MIR137, of the nine loci with genome-wide significant support either in stage 1 or in the combined set (six loci, 2q32.3, 8p23.2, 8q21.3, 10q24.32-q24.33, 11q24.2 and 18q21.2; Table 2 and Supplementary Tables 6,7) or in a joint analysis with bipolar disorder (three genes, CACNA1C, ANK3 and ITIH3-ITIH4, described below), four genes (TCF4, CACNA1C, CSMD1 and C10orf26) have predicted MIR137 target sites according to analyses using three different prediction programs (TargetScan19, PicTar20 and miRanda21). In vitro overexpression and locked nucleic acid–mediated knockdown of MIR137 in neuronal cell line N2a leads to changes in expression levels of TCF4 protein, strongly supporting the prediction that TCF4 is a target of MIR137 (L.-H. Tsai, personal communication). Our observations suggest MIR137-mediated dysregulation as a new etiologic mechanism in schizophrenia.

The International Schizophrenia Consortium (ISC) reported evidence for a polygenic contribution to schizophrenia4. An independent family based study confirmed these results, greatly minimizing the possibility of population stratification artifact12. We reevaluated the polygenic model, dividing stage 1 samples into independent training and testing sets (Supplementary Note). The training set had 15,429 subjects (over twice the size of the ISC training set), and the testing set consisted of 6,428 individuals independent of the ISC report. The proportion of variance (Nagelkerke’s r2) explained in the testing set increased from 3% in the ISC to around 6% here (Supplementary Table 9 and Supplementary Fig. 6). This estimate is much lower than the true total variation in liability that is tagged by all SNPs because SNP effects are estimated with error3,4,2225. The polygenic model appears to explain a substantial fraction of the heritability of schizophrenia4, as has been shown for other complex traits3,2628. Some of these additional risk loci are likely contained near the most highly significant results of our stage 1 analysis. Supporting this hypothesis, of the top loci that did not reach genome-wide significance in the combined stage 1 and 2 analysis, a sign test (P < 10−4) and a Fisher’s combined test (P < 10−5) both showed an excess of same-direction allelic association (41 of 51 non-MHC SNPs) in the discovery and replication datasets.

Clinical, epidemiological and genetic findings suggest shared risk factors between bipolar disorder and schizophrenia29. In stage 1, three genes with strong support had prior genome-wide significant associations with bipolar disorder: CACNA1C, the region containing ITIH3-ITIH4 (encoding inter-α (globulin) inhibitors H3 and H4) and ANK3 (encoding ankyrin 3, node of Ranvier (ankyrin G))10,11,30 (Supplementary Table 10). We performed a joint analysis with the Schizophrenia Psychiatric Genome-Wide Association Study (GWAS) Consortium (PGC) for bipolar disorder applying identical analytical methods. After removing duplicate subjects, we analyzed 16,374 cases with schizophrenia, schizoaffective disorder or bipolar disorder and 14,044 controls. Support for shared susceptibility was strengthened (Supplementary Table 11) at CACNA1C (rs4765905, P = 7.0 × 10−9), ANK3 (rs10994359, P = 2.5 × 10−8) and the ITIH3-ITIH4 region (rs2239547, P = 7.8 × 10−9), each of which reached genome-wide significance. A coding variant in ITIH4 (p.Pro698Thr; rs4687657) is in perfect LD with the most associated SNP. Although we included all subjects from an earlier report10, the increased support found with additional independent cases (N = 11,987) and controls (N = 7,835) provides further evidence for shared risk effects of schizophrenia and bipolar disorder.

The risk variants implicated here confer small risks (odds ratios ~1.10), but the polygenic analysis shows many more susceptibility variants with effects for which our sample is underpowered (Supplementary Table 12). At every stage where samples were added, we found an increase in the number of genome-wide significant loci and enhancement of signals at CACNA1C, ANK3 and ITIH3-ITIH4 when schizophrenia and bipolar disorder were jointly analyzed. Thus, gains in power offset any penalty for increased heterogeneity.

In summary, we report seven genome-wide significant schizophrenia associations (five of which are new) in a two-stage analysis of 51,695 individuals. We also report loci that confer susceptibility to both bipolar disorder and schizophrenia. The association near MIR137, associations in multiple predicted MIR137 targets and the known role of MIR137 in neuronal maturation and function together suggest an intriguing new insight into the pathogenesis of schizophrenia.

URLs

PLINK, http://pngu.mgh.harvard.edu/~purcell/plink/; Haploview, http://www.broadinstitute.org/scientific-community/science/programs/medical-and-population-genetics/haploview/haploview.

METHODS

Methods and any associated references are available in the online version of the paper at http://www.nature.com/naturegenetics/.

Supplementary Material

Supplementary Note
Online Methods

Acknowledgments

We thank the study participants and the research staff at the many study sites. Over 40 US National Institutes of Health grants and similar numbers of government grants from other countries, along with substantial private and foundation support, enabled this work. We greatly appreciate the sustained efforts of T. Lehner (National Institute of Mental Health) on behalf of the Schizophrenia Psychiatric Genome-Wide Association Study (GWAS) Consortium (PGC). Detailed acknowledgments, including grant support, are listed in the

Stephan Ripke1, Alan R Sanders2,3, Kenneth S Kendler46, Douglas F Levinson7, Pamela Sklar1,8, Peter A Holmans9,10, Dan-Yu Lin11, Jubao Duan2,3, Roel A Ophoff1215, Ole A Andreassen16,17, Edward Scolnick18, Sven Cichon1921, David St. Clair22, Aiden Corvin23, Hugh Gurling24, Thomas Werge25, Dan Rujescu26, Douglas H R Blackwood27, Carlos N Pato28, Anil K Malhotra2931, Shaun Purcell18, Frank Dudbridge32, Benjamin M Neale18, Lizzy Rossin1, Peter M Visscher33, Danielle Posthuma34,35, Douglas M Ruderfer1, Ayman Fanous5,36,37, Hreinn Stefansson38, Stacy Steinberg38, Bryan J Mowry39,40, Vera Golimbet41, Marc De Hert42, Erik G Jönsson43, István Bitter44, Olli P H Pietiläinen45,46, David A Collier47, Sarah Tosato48, Ingrid Agartz16,49, Margot Albus50, Madeline Alexander7, Richard L Amdur36,37, Farooq Amin51,52, Nicholas Bass24, Sarah E Bergen1, Donald W Black53, Anders D Børglum54,55, Matthew A Brown56, Richard Bruggeman57, Nancy G Buccola58, William F Byerley59,60, Wiepke Cahn61, Rita M Cantor14,15, Vaughan J Carr62, Stanley V Catts63, Khalid Choudhury24, C Robert Cloninger64, Paul Cormican23, Nicholas Craddock9,10, Patrick A Danoy56, Susmita Datta24, Lieuwe de Haan65, Ditte Demontis54, Dimitris Dikeos66, Srdjan Djurovic16,67, Peter Donnelly68,69, Gary Donohoe23, Linh Duong25, Sarah Dwyer9,10, Anders Fink-Jensen70, Robert Freedman71, Nelson B Freimer14, Marion Friedl26, Lyudmila Georgieva9,10, Ina Giegling26, Michael Gill23, Birte Glenthøj72, Stephanie Godard73, Marian Hamshere9,10, Mark Hansen74, Thomas Hansen25, Annette M Hartmann26, Frans A Henskens75, David M Hougaard76, Christina M Hultman77, Andrés Ingason25, Assen V Jablensky78, Klaus D Jakobsen25, Maurice Jay79,132, Gesche Jürgens80, René S Kahn61, Matthew C Keller81, Gunter Kenis82, Elaine Kenny23, Yunjung Kim83, George K Kirov9,10, Heike Konnerth26, Bettina Konte26, Lydia Krabbendam84, Robert Krasucki24, Virginia K Lasseter85,132, Claudine Laurent79, Jacob Lawrence24, Todd Lencz2931, F Bernard Lerer86, Kung-Yee Liang87, Paul Lichtenstein77, Jeffrey A Lieberman88, Don H Linszen65, Jouko Lönnqvist89, Carmel M Loughland90, Alan W Maclean27, Brion S Maher46, Wolfgang Maier91, Jacques Mallet92, Pat Malloy27, Manuel Mattheisen19,21,93, Morten Mattingsdal16,94, Kevin A McGhee27, John J McGrath39,40, Andrew McIntosh27, Duncan E McLean95, Andrew McQuillin24, Ingrid Melle16,17, Patricia T Michie96,97, Vihra Milanova98, Derek W Morris23, Ole Mors55, Preben B Mortensen99, Valentina Moskvina9,10, Pierandrea Muglia100,101, Inez Myin-Germeys84, Deborah A Nertney39,40, Gerald Nestadt85, Jimmi Nielsen102, Ivan Nikolov9,10, Merete Nordentoft103, Nadine Norton9,10, Markus M Nöthen19,21, Colm T O’Dushlaine23, Ann Olincy71, Line Olsen25, F Anthony O’Neill104, Torben F Ørntoft105,106, Michael J Owen9,10, Christos Pantelis107, George Papadimitriou66, Michele T Pato28, Leena Peltonen45,46,108,132, Hannes Petursson109, Ben Pickard110, Jonathan Pimm24, Ann E Pulver85, Vinay Puri24, Digby Quested111, Emma M Quinn23, Henrik B Rasmussen25, János M Réthelyi44, Robert Ribble46, Marcella Rietschel91,112, Brien P Riley46, Mirella Ruggeri48, Ulrich Schall97,113, Thomas G Schulze112,114, Sibylle G Schwab115117, Rodney J Scott118, Jianxin Shi119, Engilbert Sigurdsson109,120, Jeremy M Silverman8,121, Chris C A Spencer68, Kari Stefansson38, Amy Strange68, Eric Strengman12,13, T Scott Stroup88, Jaana Suvisaari89, Lars Terenius43, Srinivasa Thirumalai122, Johan H Thygesen25, Sally Timm123, Draga Toncheva124, Edwin van den Oord125, Jim van Os84, Ruud van Winkel42,82, Jan Veldink126, Dermot Walsh127, August G Wang128, Durk Wiersma57, Dieter B Wildenauer115,129, Hywel J Williams9,10, Nigel M Williams9,10, Brandon Wormley46, Stan Zammit9,10, Patrick F Sullivan77,83,130,131, Michael C O’Donovan9,10, Mark J Daly1 & Pablo V Gejman2,3

Footnotes

Note: Supplementary information is available on the Nature Genetics website.

AUTHOR CONTRIBUTIONS

The Schizophrenia Psychiatric Genome-Wide Association Study (GWAS) Consortium (PGC): overall coordination: P.V.G. Coordination of statistical analyses: M.J.D. Coordination of phenotypic analyses: K.S.K. Statistical analyses: S.R., M.J.D., P.A.H., D.-Y.L., S.P., F.D., B.M.N., L.R., P.M.V., D.P., D.M.R. Manuscript preparation: P.V.G. (primary), M.J.D. (primary), A.R.S. (primary), S.R. (primary), M.C.O. (primary), K.S.K., D.F.L., P.S., P.A.H., P.F.S. (primary), D.-Y.L., J.D., R.A.O., O.A.A., E. Scolnick. Phenotypic analyses: K.S.K., A.F., A.C., R.L.A. Stage 1 GWAS sample 1–Cardiff, UK: M.C.O., N.C., P.A.H., M. Hamshere, H.J.W., V. Moskvina, S. Dwyer, L.G., S.Z., M.J.O. Stage 1 GWAS sample 2–Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE): P.F.S., D.-Y.L., E.v.d.O., Y.K., T.S.S., J.A.L. Stage 1 GWAS sample 3–International Schizophrenia Consortium (ISC)–Aberdeen: D.St.C. Stage 1 GWAS sample 4–ISC–Cardiff: G.K.K., M.C.O., P.A.H., L.G., I.N., H.J.W., D.T., V. Milanova, M.J.O. Stage 1 GWAS sample 5–ISC–Dublin: D.W.M., C.T.O., E.K., E.M.Q., M.G., A.C. Stage 1 GWAS sample 6–ISC–Edinburgh: D.H.R.B., K.A.M., B.P., P. Malloy, A.W.M., A. McIntosh. Stage 1 GWAS sample 7–ISC–London: A. McQuillin, K.C., S. Datta, J.P., S. Thirumalai, V.P., R.K., J. Lawrence, D.Q., N.B., H.G. Stage 1 GWAS sample 8–ISC–Portugal: M.T.P., C.N.P., A.F. Stage 1 GWAS sample 9–ISC–SW1–Sweden, stage 1 GWAS sample 10–ISC–SW2–Sweden, stage 2 replication follow-up sample 16–SW3–Sweden, stage 2 replication follow-up sample 17–SW4–Sweden: C.M.H., P.L., S.E.B., S.P., E. Scolnick, P.S., P.F.S. Stage 1 GWAS sample 11–Molecular Genetics of Schizophrenia (MGS): J. Shi, D.F.L., J.D., A.R.S., M.C.K., B.J.M., A.O., F.A., C.R.C., J.M.S., N.G.B., W.FB., D.W.B., K.S.K., R.F., P.V.G. Stage 1 GWAS sample 12–Schizophrenia Genetics Consortium (SGENE)–Bonn: S.C., M. Rietschel, M.M.N., W.M., T.G.S., M. Mattheisen. Stage 1 GWAS sample 13–SGENE–Copenhagen, stage 2 replication follow-up sample 5–SGENE–Copenhagen: T.H., A.I., K.D.J., L.D., G.J., H.B.R., B.G., J.N., S. Timm, L.O., A.G.W., A.F.-J., J.H.T., T.W. Stage 1 GWAS sample 14–SGENE–Munich, stage 2 replication follow-up sample 12–SGENE–Munich, stage 2 replication follow-up sample 13–SGENE–Munich: I.G., A.M.H., H.K., M.F., B.K., P. Muglia, D.R. Stage 1 GWAS sample 15–SGENE–Thematic Organized Psychoses Research 3 (TOP3): S. Djurovic, M. Mattingsdal, I.A., I.M., O.A.A. Stage 1 GWAS sample 16–SGENE–UCLA: R.A.O., R.M.C., N.B.F., R.S.K., D.H.L., J.v.O., D. Wiersma, R.B., W.C., L.d.H., L.K., I.M.-G., E. Strengman. Stage 1 GWAS sample 17–Zucker Hillside: A.K.M., T.L. Stage 2 replication follow-up sample 1–multicenter pedigree: P.A.H., B.P.R., A.E.P., M.J.O., D.B.W., P.V.G., B.J.M., C.L., K.S.K., G.N., N.M.W., S.G.S., A.R.S., M. Hansen, D.A.N., J.M., B.W., V.K.L., M.C.O., J.D., M. Albus, M. Alexander, S.G., R.R., K.-Y.L., N.N., W.M., G.P., D. Walsh, M.J., F.A.O., F.B.L., D. Dikeos, J.M.S., D.F.L. Stage 2 replication follow-up sample 2–SGENE–Aarhus: A.D.B., D. Demontis, P.B.M., D.M.H., T.F.Ø., O.M. Stage 2 replication follow-up sample 3–SGENE–Aarhus: O.M., M.N., A.D.B. Stage 2 replication follow-up sample 4–SGENE–Belgium: R.v.W., G.K., M.D.H., J.V. Stage 2 replication follow-up sample 6–SGENE–Iceland: H.S., S.S., E. Sigurdsson, H.P., K.S. Stage 2 replication follow-up sample 7–SGENE–England: D.A.C. Stage 2 replication follow-up sample 8–SGENE–Helsinki, stage 2 replication follow-up sample 11–SGENE–Kuusamo: L.P., O.P.H.P., J. Suvisaari, J. Lönnqvist. Stage 2 replication follow-up sample 9–SGENE–Hungary: I.B., J.M.R. Stage 2 replication follow-up sample 10–SGENE–Italy: M. Ruggeri, S. Tosato. Stage 2 replication follow-up sample 14–SGENE–Russia: V.G. Stage 2 replication follow-up sample 15–SGENE–Sweden: E.G.J., I.A., L.T. Stage 2 replication follow-up sample 18–University of Queensland: B.J.M., M.A.B., P.A.D., J.J.M., D.E.M. Stage 2 replication follow-up sample 18–Australian Schizophrenia Research Bank: B.J.M., V.J.C., R.J.S., S.V.C., F.A.H., A.V.J., C.M.L., P.T.M., C.P., U.S. Stage 2 replication follow-up sample 19–Irish Schizophrenia Genomics Consortium (ISGC): A.C., D.W.M., P.C., B.S.M., C.T.O., G.D., F.A.O., M.G., K.S.K., B.P.R., ISGC (see the Acknowledgments in the Supplementary Note for additional contributors not listed above). Stage 2 replication follow-up sample 19–Wellcome Trust Case Control Consortium 2 (WTCCC2): P.D. (Chair of Management Committee; Data and Analysis Group), C.C.A.S. (Data and Analysis Group; Publications Committee), A.S. (Data and Analysis Group), WTCCC2 (see Acknowledgments in the Supplementary Note for additional contributors not listed above). All authors contributed to the current version of the paper.

COMPETING FINANCIAL INTERESTS

The authors declare competing financial interests: details accompany the full-text HTML version of the paper at http://www.nature.com/naturegenetics/.

Reprints and permissions information is available online at http://www.nature.com/reprints/index.html.

1

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

2

Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston, Illinois, USA.

3

Department of Psychiatry and Behavioral Sciences, University of Chicago, Chicago, Illinois, USA.

4

Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.

5

Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.

6

Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.

7

Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, USA.

8

Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, USA.

9

Medical Research Council (MRC) Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK.

10

Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, UK.

11

Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA.

12

Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands.

13

Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands.

14

University of California at Los Angeles (UCLA) Center for Neurobehavioral Genetics, University of California at Los Angeles, Los Angeles, California, USA.

15

Department of Human Genetics, University of California at Los Angeles, Los Angeles, California, USA.

16

Psychiatry Section, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.

17

Department of Psychiatry, Oslo University Hospital, Oslo, Norway.

18

Broad Institute, Cambridge, Massachusetts, USA.

19

Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany.

20

Institute of Neuroscience and Medicine (INM-1), Research Center Juelich, Juelich, Germany.

21

Institute of Human Genetics, University of Bonn, Bonn, Germany.

22

Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK.

23

Neuropsychiatric Genetics Research Group, Trinity College Dublin, Dublin, Ireland.

24

Molecular Psychiatry Laboratory, Research Department of Mental Health Sciences, University College London Medical School, Windeyer Institute of Medical Sciences, London, UK.

25

Institute of Biological Psychiatry, Mental Health Center (MHC) Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark.

26

Molecular and Clinical Neurobiology, Department of Psychiatry, Ludwig-Maximilians-University, Munich, Germany.

27

Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK.

28

Keck School of Medicine, University of Southern California, Los Angeles, California, USA.

29

Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of the North Shore-Long Island Jewish Health System, Glen Oaks, New York, USA.

30

Center for Psychiatric Neuroscience, The Feinstein Institute for Medical Research, Manhasset, New York, USA.

31

Department of Psychiatry and Behavioral Science, Albert Einstein College of Medicine of Yeshiva University, New York, New York, USA.

32

Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.

33

Queensland Statistical Genetics Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland, Australia.

34

Vrije Universiteit (VU), Center for Neurogenomics and Cognitive Research (CNCR), Department of Functional Genomics, Amsterdam, The Netherlands.

35

VU Medical Centre, Department of Medical Genomics, Amsterdam, The Netherlands.

36

Washington Veteran’s Affairs Medical Center, Washington, DC, USA.

37

Department of Psychiatry, Georgetown University School of Medicine, Washington, DC, USA.

38

deCODE Genetics, Reykjavik, Iceland.

39

Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.

40

Queensland Centre for Mental Health Research, University of Queensland, Brisbane, Queensland, Australia.

41

Mental Health Research Center, Russian Academy of Medical Sciences, Moscow, Russia.

42

University Psychiatric Centre, Catholic University Leuven, Kortenberg, Belgium.

43

Department of Clinical Neuroscience, Human Brain Informatics (HUBIN) Project, Karolinska Institutet and Hospital, Stockholm, Sweden.

44

Semmelweis University, Department of Psychiatry and Psychotherapy, Budapest, Hungary.

45

Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.

46

Department of Medical Genetics, University of Helsinki, Helsinki, Finland.

47

Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King’s College, London, UK.

48

Section of Psychiatry and Clinical Psychology, University of Verona, Verona, Italy.

49

Department of Research, Diakonhjemmet Hospital, Oslo, Norway.

50

State Mental Hospital, Haar, Germany.

51

Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia, USA.

52

Department of Psychiatry and Behavioral Sciences, Atlanta Veterans Affairs Medical Center, Atlanta, Georgia, USA.

53

Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.

54

Institute of Human Genetics, University of Aarhus, Aarhus, Denmark.

55

Centre for Psychiatric Research, Aarhus University Hospital, Risskov, Denmark.

56

University of Queensland Diamantina Institute, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia.

57

University Medical Center Groningen, Department of Psychiatry, University of Groningen, Groningen, The Netherlands.

58

School of Nursing, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.

59

Department of Psychiatry, University of California at San Francisco, San Francisco, California, USA.

60

NCIRE (Northern California Institute for Research and Education), San Francisco, California, USA.

61

Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands.

62

School of Psychiatry, University of New South Wales and Schizophrenia Research Institute, Sydney, New South Wales, Australia.

63

Department of Psychiatry, University of Queensland, Royal Brisbane Hospital, Brisbane, Australia.

64

Department of Psychiatry, Washington University, St. Louis, Missouri, USA.

65

Academic Medical Centre, University of Amsterdam, Department of Psychiatry, Amsterdam, The Netherlands.

66

Department of Psychiatry, University of Athens Medical School, Athens, Greece.

67

Department of Medical Genetics, Oslo University Hospital, Oslo, Norway.

68

Wellcome Trust Centre for Human Genetics, Oxford, UK.

69

Department of Statistics, University of Oxford, Oxford, UK.

70

Mental Health Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark.

71

Department of Psychiatry, University of Colorado Denver, Aurora, Colorado, USA.

72

Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, Copenhagen University Hospital, Glostrup, Denmark.

73

INSERM, Institut de Myologie, Hôpital de la Pitié-Salpêtrière, Paris, France.

74

Illumina, Inc., La Jolla, California, USA.

75

School of Electrical Engineering and Computing Science, University of Newcastle, Newcastle, New South Wales, Australia.

76

Section of Neonatal Screening and Hormones, Department of Clinical Chemistry and Immunology, The State Serum Institute, Copenhagen, Denmark.

77

Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.

78

Centre for Clinical Research in Neuropsychiatry, School of Psychiatry and Clinical Neurosciences, The University of Western Australia, Perth, Western Australia, Australia.

79

Department of Child and Adolescent Psychiatry, Pierre and Marie Curie Faculty of Medicine, Paris, France.

80

Department of Clinical Pharmacology, Bispebjerg University Hospital, Copenhagen, Denmark.

81

Department of Psychology, University of Colorado, Boulder, Boulder, Colorado, USA.

82

Department of Psychiatry and Psychology, School of Mental Health and Neuroscience, European Graduate School of Neuroscience (EURON), South Limburg Mental Health Research and Teaching Network (SEARCH), Maastricht University Medical Centre, Maastricht, The Netherlands.

83

Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.

84

Maastricht University Medical Centre, South Limburg Mental Health Research and Teaching Network, Maastricht, The Netherlands.

85

Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.

86

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

87

Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA.

88

Department of Psychiatry, Columbia University, New York, New York, USA.

89

Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland.

90

Schizophrenia Research Institute, Sydney and Centre for Brain and Mental Health Research, University of Newcastle, Newcastle, New South Wales, Australia.

91

Department of Psychiatry, University of Bonn, Bonn, Germany.

92

Laboratoire de Génétique Moléculaire de la Neurotransmission et des Processus Neurodégénératifs, Centre National de la Recherche Scientifique, Hôpital de la Pitié Salpêtrière, Paris, France.

93

Institute of Medical Biometry, Informatics and Epidemiology (IMBIE), University of Bonn, Bonn, Germany.

94

Department of Research, Sørlandet Hospital, Kristiansand, Norway.

95

Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, Queensland, Australia.

96

Functional NeuroImaging Laboratory, School of Psychology, University of Newcastle, Sydney, New South Wales, Australia.

97

Schizophrenia Research Institute, Sydney, New South Wales, Australia.

98

Department of Psychiatry, First Psychiatric Clinic, Alexander University Hospital, Sofia, Bulgaria.

99

National Centre for Register-Based Research, University of Aarhus, Aarhus, Denmark.

100

Department of Psychiatry, University of Toronto, Toronto, Canada.

101

NeuroSearch A/S, Ballerup, Denmark.

102

Unit for Psychiatric Research, Aalborg Psychiatric Hospital, Aalborg, Denmark.

103

Psychiatric Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark.

104

Department of Psychiatry, Queens University, Belfast, Ireland.

105

ARoS Applied Biotechnology A/S, Skejby, Denmark.

106

Department of Molecular Medicine, Aarhus University Hospital, Skejby, Denmark.

107

Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Melbourne, Victoria, Australia.

108

Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK.

109

Department of Psychiatry, National University Hospital, Reykjavik, Iceland.

110

Strathclyde Institute of Pharmacy and Biomedical Sciences, The John Arbuthnott Building, University of Strathclyde, Glasgow, UK.

111

Department of Psychiatry, University of Oxford, Warneford Hospital, Headington, Oxford, UK.

112

Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany.

113

Priority Centre for Brain and Mental Health Research, University of Newcastle, Sydney, New South Wales, Australia.

114

Department of Psychiatry and Psychotherapy, Georg-August-University, Göttingen, Germany.

115

School of Psychiatry and Clinical Neurosciences, University of Western Australia, Perth, Western Australia, Australia.

116

Department of Psychiatry, University of Erlangen-Nuremberg, Erlangen, Germany.

117

Centre for Medical Research, Western Australian Institute for Medical Research, University of Western Australia, Perth, Western Australia, Australia.

118

Centre for Information Based Medicine, University of Newcastle, Hunter Medical Research Institute, Newcastle and Schizophrenia Research Institute, Sydney, New South Wales, Australia.

119

Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA.

120

Department of Psychiatry, University of Iceland, Reykjavik, Iceland.

121

Department of Psychiatry, Veterans Affairs Medical Center, New York, New York, USA.

122

West Berkshire National Health Service (NHS) Trust, Reading, UK.

123

Mental Health Center Frederiksberg, Copenhagen University Hospital, Copenhagen, Denmark.

124

Department of Medical Genetics, University Hospital Maichin Dom, Sofia, Bulgaria.

125

Department of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, USA.

126

Rudolf Magnus Institute of Neuroscience, Department of Neurology, Universitair Medisch Centrum (UMC) Utrecht, Utrecht, The Netherlands.

127

The Health Research Board, Dublin, Ireland.

128

Mental Health Center Amager, Copenhagen University Hospital, Copenhagen, Denmark.

129

Centre for Clinical Research in Neuropsychiatry, Graylands Hospital, Mt Claremont, Western Australia, Australia.

130

Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.

131

Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.

132

Deceased. Correspondence should be addressed to P.V.G. (pgejman@gmail.com).

References

  • 1.Altshuler DM, et al. Integrating common and rare genetic variation in diverse human populations. Nature. 2010;467:52–58. doi: 10.1038/nature09298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Barrett JC, et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn’s disease. Nat Genet. 2008;40:955–962. doi: 10.1038/NG.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lango Allen H, et al. Hundreds of variants clustered in genomic loci and biological pathways affect human height. Nature. 2010;467:832–838. doi: 10.1038/nature09410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Purcell SM, et al. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature. 2009;460:748–752. doi: 10.1038/nature08185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Price AL, et al. Discerning the ancestry of European Americans in genetic association studies. PLoS Genet. 2008;4:e236. doi: 10.1371/journal.pgen.0030236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Devlin B, Roeder K. Genomic control for association studies. Biometrics. 1999;55:997–1004. doi: 10.1111/j.0006-341x.1999.00997.x. [DOI] [PubMed] [Google Scholar]
  • 7.Dudbridge F, Gusnanto A. Estimation of significance thresholds for genomewide association scans. Genet Epidemiol. 2008;32:227–234. doi: 10.1002/gepi.20297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Stefansson H, et al. Common variants conferring risk of schizophrenia. Nature. 2009;460:744–747. doi: 10.1038/nature08186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Shi J, et al. Common variants on chromosome 6p22.1 are associated with schizophrenia. Nature. 2009;460:753–757. doi: 10.1038/nature08192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ferreira MA, et al. Collaborative genome-wide association analysis supports a role for ANK3 and CACNA1C in bipolar disorder. Nat Genet. 2008;40:1056–1058. doi: 10.1038/ng.209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Green EK, et al. The bipolar disorder risk allele at CACNA1C also confers risk of recurrent major depression and of schizophrenia. Mol Psychiatry. 2010;15:1016–1022. doi: 10.1038/mp.2009.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ruderfer DM, et al. A family-based study of common polygenic variation and risk of schizophrenia. Mol Psychiatry. 2011 April 12; doi: 10.1038/mp.2011.34. published online. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bemis LT, et al. MicroRNA-137 targets microphthalmia-associated transcription factor in melanoma cell lines. Cancer Res. 2008;68:1362–1368. doi: 10.1158/0008-5472.CAN-07-2912. [DOI] [PubMed] [Google Scholar]
  • 14.Srikantan V, et al. PCGEM1, a prostate-specific gene, is overexpressed in prostate cancer. Proc Natl Acad SciUSA. 2000;97:12216–12221. doi: 10.1073/pnas.97.22.12216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Szulwach KE, et al. Cross talk between microRNA and epigenetic regulation in adult neurogenesis. J Cell Biol. 2010;189:127–141. doi: 10.1083/jcb.200908151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Silber J, et al. miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells. BMC Med. 2008;6:14. doi: 10.1186/1741-7015-6-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Smrt RD, et al. MicroRNA miR-137 regulates neuronal maturation by targeting ubiquitin ligase mind bomb-1. Stem Cells. 2010;28:1060–1070. doi: 10.1002/stem.431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Potkin SG, et al. Identifying gene regulatory networks in schizophrenia. Neuroimage. 2010;53:839–847. doi: 10.1016/j.neuroimage.2010.06.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120:15–20. doi: 10.1016/j.cell.2004.12.035. [DOI] [PubMed] [Google Scholar]
  • 20.Krek A, et al. Combinatorial microRNA target predictions. Nat Genet. 2005;37:495–500. doi: 10.1038/ng1536. [DOI] [PubMed] [Google Scholar]
  • 21.John B, et al. Human MicroRNA targets. PLoS Biol. 2004;2:e363. doi: 10.1371/journal.pbio.0020363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Yang J, et al. Common SNPs explain a large proportion of the heritability for human height. Nat Genet. 2010;42:565–569. doi: 10.1038/ng.608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Yang J, et al. Genomic inflation factors under polygenic inheritance. Eur J Hum Genet. 2011;19:807–812. doi: 10.1038/ejhg.2011.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Visscher PM, Yang J, Goddard ME. A commentary on ‘common SNPs explain a large proportion of the heritability for human height’ by Yang et al. (2010) Twin Res Hum Genet. 2010;13:517–524. doi: 10.1375/twin.13.6.517. [DOI] [PubMed] [Google Scholar]
  • 25.Lee SH, Wray NR, Goddard ME, Visscher PM. Estimating missing heritability for disease from genome-wide association studies. Am J Hum Genet. 2011;88:294–305. doi: 10.1016/j.ajhg.2011.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Speliotes EK, et al. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat Genet. 2010;42:937–948. doi: 10.1038/ng.686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wei Z, et al. From disease association to risk assessment: an optimistic view from genome-wide association studies on type 1 diabetes. PLoS Genet. 2009;5:e1000678. doi: 10.1371/journal.pgen.1000678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bush WS, et al. Evidence for polygenic susceptibility to multiple sclerosis—the shape of things to come. Am J Hum Genet. 2010;86:621–625. doi: 10.1016/j.ajhg.2010.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lichtenstein P, et al. Common genetic determinants of schizophrenia and bipolar disorder in Swedish families: a population-based study. Lancet. 2009;373:234–239. doi: 10.1016/S0140-6736(09)60072-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Scott LJ, et al. Genome-wide association and meta-analysis of bipolar disorder in individuals of European ancestry. Proc Natl Acad Sci USA. 2009;106:7501–7506. doi: 10.1073/pnas.0813386106. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Note
Online Methods

RESOURCES