Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Exome sequencing of Japanese schizophrenia multiplex families supports the involvement of calcium ion channels

  • Miho Toyama,

    Roles Data curation, Formal analysis, Investigation, Validation, Visualization, Writing – original draft

    Affiliation Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan

  • Yuto Takasaki,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Resources

    Affiliation Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan

  • Aleksic Branko,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Project administration, Supervision, Validation, Writing – original draft, Writing – review & editing

    Affiliation Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan

  • Hiroki Kimura ,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Resources, Validation, Writing – review & editing

    kimura.hiroki@med.nagoya-u.ac.jp

    Affiliation Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan

  • Hidekazu Kato,

    Roles Investigation, Supervision, Validation, Writing – review & editing

    Affiliation Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan

  • Yoshihiro Nawa,

    Roles Investigation, Validation, Writing – review & editing

    Affiliation Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan

  • Itaru Kushima,

    Roles Conceptualization, Project administration, Supervision

    Affiliation Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan

  • Kanako Ishizuka,

    Roles Conceptualization, Resources, Supervision, Writing – review & editing

    Affiliation Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan

  • Teppei Shimamura,

    Roles Methodology, Supervision, Writing – review & editing

    Affiliation Division of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan

  • Tomoo Ogi,

    Roles Methodology, Supervision, Writing – review & editing

    Affiliations Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan, Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan

  • Norio Ozaki

    Roles Conceptualization, Funding acquisition, Project administration, Resources, Supervision, Writing – review & editing

    Affiliations Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan, Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan

Abstract

Background

Most sequencing studies of schizophrenia (SCZ) have focused on de novo genetic variants due to interpretability. However, investigating shared rare variants among patients in the same multiplex family is also important. Relatively large-scale analyses of SCZ multiplex families have been done in Caucasian populations, but whether detected variants are also pathogenic in the Japanese population is unclear because of ethnic differences in rare variants.

Materials and methods

We performed whole-exome sequencing (WES) of 14 Japanese SCZ multiplex families. After quality control and filtering, we identified rare variants shared among affected persons within the same family. A gene ontology (GO) analysis was performed to identify gene categories possibly affected by these candidate variants.

Results

We found 530 variants in 486 genes as potential candidate variants from the 14 SCZ multiplex families examined. The GO analysis demonstrated significant enrichment in calcium channel activity.

Conclusion

This study provides supporting evidence that calcium ion channel activity is involved in SCZ. WES of multiplex families is a potential means of identifying disease-associated rare variants for SCZ.

Introduction

Schizophrenia (SCZ) is a chronic and severe mental disorder characterized by some combination of hallucinations, delusions, and extremely disordered thinking and behavior that impairs daily functioning [1]. The lifetime prevalence of SCZ is 0.3–0.7%, and the standardized mortality ratio is 2.5 [2, 3]. Both genetic and environmental factors affect the risk for SCZ [4, 5]. Population-based and twin concordance studies indicated that the heritability of SCZ is 60–80%, with only subtle contributions from environmental factors [6, 7].

As genetic factors play a significant role in the etiology of SCZ, many genetic studies of the disease have been conducted. Linkage studies identified multiple genetic risk loci for SCZ; however, these studies were not sufficient to suggest specific causative genes [8]. Genome-wide association studies (GWASs) have identified numerous loci significantly associated with SCZ [9, 10] based on the common disease–common variant hypothesis. However, the effect sizes of the individual single-nucleotide polymorphisms identified in GWASs are too small to explain the high heritability of SCZ demonstrated in cohort studies, which has been designated “missing heritability” [11]. To overcome this problem, it is necessary to consider rare variants such as single-nucleotide variants (SNVs) or copy number variants [12].

The feasibility of exploring disease-associated SNVs has been enhanced due to the advent of whole-genome sequencing (WGS) and whole-exome sequencing (WES) techniques, with the latter providing a more rapid and cost-effective approach for sequencing protein-coding regions across the genome. However, efficiently extracting candidates from a large number of detected mutations is challenging. As such, most of these studies have focused on de novo genetic variants or rare variants significantly associated with SCZ due to interpretability [1316] in terms of their large estimated effect sizes and the possibility of functional validation.

In addition, patients in families with multiple affected members are likely to be enriched in genetic factors that strongly affect the development of SCZ [17]. Therefore, it is also essential to investigate variants shared among patients in order to elucidate the association between transmitted variants and SCZ. Indeed, several sequencing studies have focused on shared variants associated with bipolar disorder [18, 19] and autism spectrum disorder [20, 21] in patients in the same multiplex family. A previous WES study of SCZ multiplex families demonstrated that several loci could potentially affect synaptic plasticity and neurocognitive performance [22] and that variants in genes related to metabotropic glutamate receptor 5 (mGlu5) are more common in affected family members [23].

However, the above-mentioned relatively large-scale sequencing studies have been done in Caucasian populations. The total genetic variation of the Japanese population, however, is considered relatively low [24] compared to that of ethnically diverse populations, in particular Europeans, which can be beneficial in sequencing studies due to decreased allelic diversity [2527]. Furthermore, rare variants, which were targeted in this study due to their more recent origin, tend to be more geographically clustered and can be population specific, thus potentially revealing new SCZ candidates beyond those identified in studies of Caucasian populations.

We therefore hypothesized that we could identify variants associated with SCZ susceptibility in the Japanese population via analyses of Japanese SCZ multiplex families, even if the sample sizes were small compared with studies of Caucasian populations. Individual rare variants provide limited power for identifying significant trait associations, and thus, multi-variant and/or multi-genic approaches such as gene set enrichment tests are necessary.

To address this hypothesis, a WES study of multiplex families within the Japanese population was conducted to identify disease-associated rare variants or gene sets.

Materials and methods

Participants—SCZ multiplex families

A multiplex family was defined as a family having more than one member with SCZ. DNA samples were collected from peripheral blood or saliva of 29 patients with SCZ, 1 patient with obsessive-compulsive disorder (OCD), and 9 healthy individuals from 14 SCZ multiplex families in Japan (Fig 1, S1 Table). Among the 14 SCZ multiplex families, pedigree 10 was a consanguineous family. All families were unrelated, lived on the mainland of Japan, and self-identified as Japanese. All patients fulfilled the criteria for SCZ listed in the Diagnostic and Statistical Manual of Mental Disorders–Fifth Edition (DSM-5).

thumbnail
Fig 1. Sequenced samples.

Dashed lines indicate individuals who underwent WES. Squares indicate males, and circles indicate females. Shading indicates an affected individual, and slash marks indicate deceased individuals. We performed WES for 29 patients with SCZ, 1 patient with obsessive-compulsive disorder (OCD), and 9 healthy individuals from 14 SCZ multiplex families in Japan.

https://doi.org/10.1371/journal.pone.0268321.g001

The Ethics Committees of Nagoya University Graduate School of Medicine approved this study, and written informed consent was obtained from all subjects. The study was conducted in accordance with the Helsinki Declaration of 1975 and its later amendments or comparable ethical standards.

WES

The library was prepared using SureSelect XT Human All Exon V5 (Agilent Technologies). WES was performed on a HiSeq2500 sequencer (Illumina) with paired-end 100-bp reads. WES data reported are available only upon request, as the data contain potentially identifying or sensitive pedigree information. Low-quality reads were excluded using the FASTX-Toolkit, and the remaining reads were mapped to the Human 1kg Reference (GRCh37 + decoy) using the BWA-MEM algorithm. Duplicated reads were removed using Picard. Variants were called using the HaplotypeCaller in the Genome Analysis Toolkit (GATK) [28] and annotated using ANNOVAR [29] with GENCODE Comprehensive gene annotation ver. 19.

Quality control

To reduce the number of false positives, only SNVs satisfying the following criteria were included: read depth ≥10, genotyping quality ≥20, and alternative allele ratio ≥25%, which was derived from the number of reads with alternative alleles divided by the total number of reads. We also included variants passed through the GATK VQSR filter and not in segmental duplications. Furthermore, using in-house WES data for 1,781 samples, variants with ≥20 detections were excluded to avoid platform-dependent sequencing errors (i.e., false positives). We then performed analyses of relatedness using the—relatedness2 option of vcftools [30] software to confirm the prior information about each multiplex family [31].

Filtering conditions

To identify pathogenic SNVs and indels, we selected those meeting the following conditions: (1) variants that cause protein alterations; (2) variants located in the splicing site, including synonymous variants detected by ANNOVAR with GENCODE comprehensive gene annotation ver. 19., because synonymous variants that disrupt exonic splice enhancers could be a common cause of genetic disorders [32]; (3) variants with an allele frequency ≤1% in the following databases: 1000 Genome Project (2015 August), total population without psychiatric cohorts in Exome Aggregation Consortium ver. 0.3, Eastern Asian population in Exome Aggregation Consortium ver. 0.3, Human Genetic Variation Database (http://www.hgvd.genome.med.kyoto-u.ac.jp), and Japanese Multi Omics Reference Panel (https://jmorp.megabank.tohoku.ac.jp/); (4) variants shared only among patients (we also selected de novo variants only from pedigrees whose parents were unaffected, as pedigrees with affected parents could have shared rare variants between cases); and (5) variants in genes with a percentile residual variation intolerance score (RVIS) [33] (ExAC_0.05 threshold) ≤25% and genes expressed in brain regions based on data from the Human Protein Atlas with normalized expression ≥1, as we assumed that variants occurring in genes highly intolerant of protein alterations and expressed in the brain could be deleterious for biological functions.

For deleterious variants and/or de novo or homozygous variants, we manually inspected their calls using Integrated Genomics Viewer [34] ver.2.7.2 and confirmed their exonic functions (change in protein structure and/or function induced by a variant in the exon, such as synonymous, nonsynonymous, or loss of function) using Ensembl genome browser GRCh37 (http://grch37.ensembl.org/index.html).

Gene ontology (GO) analysis

We performed GO analysis (http://geneontology.org/docs/go-enrichment-analysis/) using Cytoscape and its GeneMANIA plugin [35]. To correct in multiple comparisons, Q-values derived via the Benjamini-Hochberg procedure were used to judge the significance of results. The significance level was set at Q-value <0.05.

Results

WES analysis

We performed WES for 14 SCZ multiplex families in Japan (Fig 1). The data were quality checked and filtered using the following criteria: 1) low frequency (an allele frequency ≤1% in the databases), 2) protein-altering (missense and splice site variants), and 3) variants in the intolerant genes (RVIS ≤ 25%) and genes expressed in brain regions. Finally, we identified 525 variants among 481 genes carried by at least two patients in the same family. A summary of the filtered variants is presented in S2 and S3 Tables. Loss-of-function variants among these candidate variants are presented in Table 1. In addition to the 525 variants identified among the 481 genes, we also identified a number of de novo variants (Table 2). Therefore, we identified a total of 530 variants among 486 genes as potential multiplex family candidate variants for SCZ.

Of the 14 pedigrees examined, seven (pedigrees 1, 4, 5, 10, 11, 13, and 14) included control samples. We selected 199 variants in 190 genes found in these seven pedigrees as “strict-filtered” for further analysis, as we sequenced both affected and unaffected family members in these pedigrees to exclude variants carried by healthy individuals.

GO analysis

To identify gene categories that could be affected by the identified candidate variants, we performed GO analysis. Significantly enriched categories (Q-value <0.05) are shown in Table 3. We tested the enrichment of the 199 variants identified in the 190 “strict-filtered” genes (pedigrees 1, 4, 5, 10, 11, 13, and 14) and found significant enrichment of genes associated with calcium channel activity (Q = 0.032). A similar result was also observed for the 530 variants identified in 486 genes that were derived from whole pedigrees in this study (Q = 0.011).

Discussion

This is the first report of a WES analysis of Japanese SCZ multiplex families. After WES analysis and filtering of 14 SCZ multiplex families, we selected variants shared only among patients (we also selected de novo variants only from pedigrees whose parents were unaffected, as pedigrees with affected parents could have shared rare variants between cases), and we identified a total of 530 SCZ candidate SNVs and indels among 486 genes. In an in silico analysis involving 530 SCZ candidate SNVs and indels among 486 genes, we demonstrated that many candidate variants were located in genes related to calcium ion channels that have also been reported as involved in the pathophysiology of SCZ, as demonstrated by common variants from genome-wide association analyses [10] and rare variants from whole-genome CNV/SNV analyses [16, 36]. For example, CACNA1C, which was identified as a candidate gene in this study, was also identified as a SCZ susceptibility gene in a GWAS [10] as well as exome [16] and whole-genome CNV [36] analyses.

The 486 candidate genes from 530 SCZ candidate variants identified in our present study were enriched in calcium channel activity–related genes as the most enriched GO terms (Table 3), although there could have been enrichment of calcium channel–related GO terms among the top 25% RVISs and brain-expressed genes. Voltage-gated calcium channels are widely distributed in all parts of the brain. They are critical for mediating intracellular Ca2+ influx, which results in transmitter release from pre-synaptic endings, thereby affecting neuronal excitability and synaptic plasticity and playing a role in neurodevelopmental disorders such as SCZ [37]. On the other hand, in this study, we did not observe any GO enrichment in the SCZ-associated genes reported in two previous WES studies of SCZ multiplex families [22, 23] such as AMACR, a gene involved in fatty acid metabolism and previously implicated in SCZ [22], and genes related to the metabotropic glutamate receptor 5 (Table 3) [23].

Among the 530 candidate variants identified, we detected a male carrier of a hemizygous variant in MAOB (located on the X chromosome) and one homozygous variant in CNTN6 in a pedigree with consanguineous marriage (Table 2). The p.P109L variant in MAOB identified in family 13 was previously reported in a male SCZ patient as being inherited from his heterozygous mother [38]. This variant may cause a change in the structure of the protein’s binding site to the mitochondrial membrane [39]. We also identified this hemizygous variant in a male SCZ patient segregated from his heterozygous mother with OCD (Fig 1). Interestingly, MAOB is suggested as being associated with OCD [40].

The homozygous SNV in CNTN6 shared among affected members of pedigree 10 with consanguineous marriage is also interesting, as several recent reports have identified CNTN6 as a candidate gene involved in neurodevelopmental disorders, including SCZ [41, 42]. SNVs in CNTN6 are significantly associated with Autism spectrum disorder in particular [43]. CNTN6 encodes contactin 6, which plays a role in neuronal cell adhesion and promotes neurite outgrowth in sensory-motor neuronal pathways [44]. The homozygous variant we identified is located in the immunoglobulin C2–type domain (IGC domain), which mediates interactions with contactin-binding partners such as protein tyrosine phosphatase receptor–gamma (Ptptg), which plays a role in the molecular basis of neurodevelopmental functions [44]. Moreover, among the 486 genes examined in the present study, three genes (RBM12, NRXN1, AKT1) are registered in the OMIM (https://www.ncbi.nlm.nih.gov/omim) database as being associated with susceptibility to SCZ.

Although our results provide some support for the calcium channel–associated hypothesis, there are still limitations to our study. First, our sample size was small for an investigation of the burden and/or transmission of rare and de novo protein-altering mutations between affected and non-affected individuals, and thus, our results could be misleading. Second, we could not evaluate clinical phenotypes, especially within unaffected families. Therefore, we could not evaluate the clinical phenotype of unaffected family members with discovered variants associated with susceptibility to SCZ, such as MAOB and CNTN6. Furthermore, in this study, we could only elucidate minimal variant information for each pedigree due to data availability constraints. In future multiplex familial studies, it will be important to evaluate in detail the relationship between variants and phenotype within the same family. Third, due to the study’s small sample size, the experiments could have generated false-positive results, and strict filtering could have led to some false-negative results. The different strategies we used in the study to narrow down candidate variants likely limited false positives, however. More importantly, SCZ is not a monogenic disorder, so filtering is not necessary to identify only a few pathogenic variants; identifying disease-associated pathways would also be a useful approach to elucidate the mechanism of the disorder.

Conclusion

In conclusion, using WES analysis of 14 Japanese multiplex families, we identified a number of rare variants segregated in SCZ patients. Our results provide support for the hypothesis that calcium channel activity is related to the development of SCZ. Analyzing a larger sample size of multiplex families could confirm these results and provide additional information regarding the aspects of this disease.

Supporting information

S1 Table. Information regarding samples used in this study.

https://doi.org/10.1371/journal.pone.0268321.s001

(DOCX)

S3 Table. Information of filtered variants in this study.

https://doi.org/10.1371/journal.pone.0268321.s003

(XLSX)

S4 Table. List of genes used in the GO analysis.

https://doi.org/10.1371/journal.pone.0268321.s004

(XLSX)

Acknowledgments

We are grateful to all of the patients and their families who contributed to this study. We thank Mami Yoshida, Kiyori Monta, Hiromi Noma, and Yukari Mitsui for technical assistance, discussions, and contributions to creating and managing the database.

References

  1. 1. Owen MJ, Sawa A, Mortensen PB. Schizophrenia. Lancet. 2016;388(10039):86–97. pmid:26777917
  2. 2. van Os J, Kapur S. Schizophrenia. Lancet. 2009;374(9690):635–45. pmid:19700006
  3. 3. Kahn RS, Sommer IE, Murray RM, Meyer-Lindenberg A, Weinberger DR, Cannon TD, et al. Schizophrenia. Nat Rev Dis Primers. 2015;1:15067. pmid:27189524
  4. 4. Kimura H, Mori D, Aleksic B, Ozaki N. Elucidation of molecular pathogenesis and drug development for psychiatric disorders from rare disease-susceptibility variants. Neurosci Res. 2020. pmid:33316300
  5. 5. Murray RM, Bhavsar V, Tripoli G, Howes O. 30 Years on: How the Neurodevelopmental Hypothesis of Schizophrenia Morphed Into the Developmental Risk Factor Model of Psychosis. Schizophr Bull. 2017;43(6):1190–6. pmid:28981842
  6. 6. Hilker R, Helenius D, Fagerlund B, Skytthe A, Christensen K, Werge TM, et al. Heritability of Schizophrenia and Schizophrenia Spectrum Based on the Nationwide Danish Twin Register. Biol Psychiatry. 2018;83(6):492–8. pmid:28987712
  7. 7. Lichtenstein P, Yip BH, Bjork C, Pawitan Y, Cannon TD, Sullivan PF, et al. Common genetic determinants of schizophrenia and bipolar disorder in Swedish families: a population-based study. Lancet. 2009;373(9659):234–9. pmid:19150704
  8. 8. Ng MY, Levinson DF, Faraone SV, Suarez BK, DeLisi LE, Arinami T, et al. Meta-analysis of 32 genome-wide linkage studies of schizophrenia. Mol Psychiatry. 2009;14(8):774–85. pmid:19349958
  9. 9. Li Z, Chen J, Yu H, He L, Xu Y, Zhang D, et al. Genome-wide association analysis identifies 30 new susceptibility loci for schizophrenia. Nat Genet. 2017;49(11):1576–83. pmid:28991256
  10. 10. Schizophrenia Working Group of the Psychiatric Genomics C. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014;511(7510):421–7. pmid:25056061
  11. 11. Sullivan PF, Daly MJ, O’Donovan M. Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat Rev Genet. 2012;13(8):537–51. pmid:22777127
  12. 12. Sanders SJ, Neale BM, Huang H, Werling DM, An JY, Dong S, et al. Whole genome sequencing in psychiatric disorders: the WGSPD consortium. Nat Neurosci. 2017;20(12):1661–8. pmid:29184211
  13. 13. Fromer M, Pocklington AJ, Kavanagh DH, Williams HJ, Dwyer S, Gormley P, et al. De novo mutations in schizophrenia implicate synaptic networks. Nature. 2014;506(7487):179–84. pmid:24463507
  14. 14. Rees E, Han J, Morgan J, Carrera N, Escott-Price V, Pocklington AJ, et al. De novo mutations identified by exome sequencing implicate rare missense variants in SLC6A1 in schizophrenia. Nat Neurosci. 2020;23(2):179–84. pmid:31932766
  15. 15. Howrigan DP, Rose SA, Samocha KE, Fromer M, Cerrato F, Chen WJ, et al. Exome sequencing in schizophrenia-affected parent-offspring trios reveals risk conferred by protein-coding de novo mutations. Nat Neurosci. 2020;23(2):185–93. pmid:31932770
  16. 16. Purcell SM, Moran JL, Fromer M, Ruderfer D, Solovieff N, Roussos P, et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature. 2014;506(7487):185–90. pmid:24463508
  17. 17. Khan FF, Melton PE, McCarthy NS, Morar B, Blangero J, Moses EK, et al. Whole genome sequencing of 91 multiplex schizophrenia families reveals increased burden of rare, exonic copy number variation in schizophrenia probands and genetic heterogeneity. Schizophr Res. 2018;197:337–45. pmid:29486958
  18. 18. Goes FS, Pirooznia M, Parla JS, Kramer M, Ghiban E, Mavruk S, et al. Exome Sequencing of Familial Bipolar Disorder. JAMA Psychiatry. 2016;73(6):590–7. pmid:27120077
  19. 19. Anjanappa RM, Nayak S, Moily NS, Manduva V, Nadella RK, Viswanath B, et al. A linkage and exome study implicates rare variants of KANK4 and CAP2 in bipolar disorder in a multiplex family. Bipolar Disord. 2020;22(1):70–8. pmid:31400178
  20. 20. Ruzzo EK, Perez-Cano L, Jung JY, Wang LK, Kashef-Haghighi D, Hartl C, et al. Inherited and De Novo Genetic Risk for Autism Impacts Shared Networks. Cell. 2019;178(4):850–66 e26. pmid:31398340
  21. 21. Toma C, Torrico B, Hervas A, Valdes-Mas R, Tristan-Noguero A, Padillo V, et al. Exome sequencing in multiplex autism families suggests a major role for heterozygous truncating mutations. Mol Psychiatry. 2014;19(7):784–90. pmid:23999528
  22. 22. Kos MZ, Carless MA, Peralta J, Curran JE, Quillen EE, Almeida M, et al. Exome sequences of multiplex, multigenerational families reveal schizophrenia risk loci with potential implications for neurocognitive performance. Am J Med Genet B Neuropsychiatr Genet. 2017;174(8):817–27. pmid:28902459
  23. 23. Timms AE, Dorschner MO, Wechsler J, Choi KY, Kirkwood R, Girirajan S, et al. Support for the N-methyl-D-aspartate receptor hypofunction hypothesis of schizophrenia from exome sequencing in multiplex families. JAMA Psychiatry. 2013;70(6):582–90. pmid:23553203
  24. 24. Yamaguchi-Kabata Y, Nakazono K, Takahashi A, Saito S, Hosono N, Kubo M, et al. Japanese population structure, based on SNP genotypes from 7003 individuals compared to other ethnic groups: effects on population-based association studies. Am J Hum Genet. 2008;83(4):445–56. pmid:18817904
  25. 25. Heutink P, Oostra BA. Gene finding in genetically isolated populations. Hum Mol Genet. 2002;11(20):2507–15. pmid:12351587
  26. 26. Bonnen PE, Pe’er I, Plenge RM, Salit J, Lowe JK, Shapero MH, et al. Evaluating potential for whole-genome studies in Kosrae, an isolated population in Micronesia. Nat Genet. 2006;38(2):214–7. pmid:16429162
  27. 27. Service S, DeYoung J, Karayiorgou M, Roos JL, Pretorious H, Bedoya G, et al. Magnitude and distribution of linkage disequilibrium in population isolates and implications for genome-wide association studies. Nat Genet. 2006;38(5):556–60. pmid:16582909
  28. 28. Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinformatics. 2013;43:11 0 1–33. pmid:25431634
  29. 29. Yang H, Wang K. Genomic variant annotation and prioritization with ANNOVAR and wANNOVAR. Nat Protoc. 2015;10(10):1556–66. pmid:26379229
  30. 30. Danecek P, Auton A, Abecasis G, Albers CA, Banks E, DePristo MA, et al. The variant call format and VCFtools. Bioinformatics. 2011;27(15):2156–8. pmid:21653522
  31. 31. Manichaikul A, Mychaleckyj JC, Rich SS, Daly K, Sale M, Chen WM. Robust relationship inference in genome-wide association studies. Bioinformatics. 2010;26(22):2867–73. pmid:20926424
  32. 32. Savisaar R, Hurst LD. Exonic splice regulation imposes strong selection at synonymous sites. Genome Res. 2018;28(10):1442–54. pmid:30143596
  33. 33. Petrovski S, Wang Q, Heinzen EL, Allen AS, Goldstein DB. Genic intolerance to functional variation and the interpretation of personal genomes. PLoS Genet. 2013;9(8):e1003709. pmid:23990802
  34. 34. Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29(1):24–6. pmid:21221095
  35. 35. Montojo J, Zuberi K, Rodriguez H, Kazi F, Wright G, Donaldson SL, et al. GeneMANIA Cytoscape plugin: fast gene function predictions on the desktop. Bioinformatics. 2010;26(22):2927–8. pmid:20926419
  36. 36. Szatkiewicz JP, Fromer M, Nonneman RJ, Ancalade N, Johnson JS, Stahl EA, et al. Characterization of Single Gene Copy Number Variants in Schizophrenia. Biol Psychiatry. 2020;87(8):736–44. pmid:31767120
  37. 37. Nanou E, Catterall WA. Calcium Channels, Synaptic Plasticity, and Neuropsychiatric Disease. Neuron. 2018;98(3):466–81. pmid:29723500
  38. 38. Piton A, Gauthier J, Hamdan FF, Lafreniere RG, Yang Y, Henrion E, et al. Systematic resequencing of X-chromosome synaptic genes in autism spectrum disorder and schizophrenia. Mol Psychiatry. 2011;16(8):867–80. pmid:20479760
  39. 39. Binda C, Newton-Vinson P, Hubalek F, Edmondson DE, Mattevi A. Structure of human monoamine oxidase B, a drug target for the treatment of neurological disorders. Nat Struct Biol. 2002;9(1):22–6. pmid:11753429
  40. 40. McGregor NW, Hemmings SMJ, Erdman L, Calmarza-Font I, Stein DJ, Lochner C. Modification of the association between early adversity and obsessive-compulsive disorder by polymorphisms in the MAOA, MAOB and COMT genes. Psychiatry Res. 2016;246:527–32. pmid:27821364
  41. 41. Hu J, Liao J, Sathanoori M, Kochmar S, Sebastian J, Yatsenko SA, et al. CNTN6 copy number variations in 14 patients: a possible candidate gene for neurodevelopmental and neuropsychiatric disorders. J Neurodev Disord. 2015;7(1):26. pmid:26257835
  42. 42. Pol-Fuster J, Canellas F, Ruiz-Guerra L, Medina-Dols A, Bisbal-Carrio B, Asensio V, et al. Familial Psychosis Associated With a Missense Mutation at MACF1 Gene Combined With the Rare Duplications DUP3p26.3 and DUP16q23.3, Affecting the CNTN6 and CDH13 Genes. Front Genet. 2021;12:622886. pmid:33897758
  43. 43. Mercati O, Huguet G, Danckaert A, Andre-Leroux G, Maruani A, Bellinzoni M, et al. CNTN6 mutations are risk factors for abnormal auditory sensory perception in autism spectrum disorders. Mol Psychiatry. 2017;22(4):625–33. pmid:27166760
  44. 44. Zuko A, Bouyain S, van der Zwaag B, Burbach JP. Contactins: structural aspects in relation to developmental functions in brain disease. Adv Protein Chem Struct Biol. 2011;84:143–80. pmid:21846565