151
|
Chen X, Lee G, Maher BS, Fanous AH, Chen J, Zhao Z, Guo A, van den Oord E, Sullivan PF, Shi J, Levinson DF, Gejman PV, Sanders A, Duan J, Owen MJ, Craddock NJ, O'Donovan MC, Blackman J, Lewis D, Kirov GK, Qin W, Schwab S, Wildenauer D, Chowdari K, Nimgaonkar V, Straub RE, Weinberger DR, O'Neill FA, Walsh D, Bronstein M, Darvasi A, Lencz T, Malhotra AK, Rujescu D, Giegling I, Werge T, Hansen T, Ingason A, Nöethen MM, Rietschel M, Cichon S, Djurovic S, Andreassen OA, Cantor RM, Ophoff R, Corvin A, Morris DW, Gill M, Pato CN, Pato MT, Macedo A, Gurling HMD, McQuillin A, Pimm J, Hultman C, Lichtenstein P, Sklar P, Purcell SM, Scolnick E, St Clair D, Blackwood DHR, Kendler KS, GROUP investigators, International Schizophrenia Consortium. GWA study data mining and independent replication identify cardiomyopathy-associated 5 (CMYA5) as a risk gene for schizophrenia. Mol Psychiatry 2011; 16:1117-29. [PMID: 20838396 PMCID: PMC3443634 DOI: 10.1038/mp.2010.96] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2010] [Revised: 08/03/2010] [Accepted: 08/11/2010] [Indexed: 12/24/2022]
Abstract
We conducted data-mining analyses using the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) and molecular genetics of schizophrenia genome-wide association study supported by the genetic association information network (MGS-GAIN) schizophrenia data sets and performed bioinformatic prioritization for all the markers with P-values ≤0.05 in both data sets. In this process, we found that in the CMYA5 gene, there were two non-synonymous markers, rs3828611 and rs10043986, showing nominal significance in both the CATIE and MGS-GAIN samples. In a combined analysis of both the CATIE and MGS-GAIN samples, rs4704591 was identified as the most significant marker in the gene. Linkage disequilibrium analyses indicated that these markers were in low LD (3 828 611-rs10043986, r(2)=0.008; rs10043986-rs4704591, r(2)=0.204). In addition, CMYA5 was reported to be physically interacting with the DTNBP1 gene, a promising candidate for schizophrenia, suggesting that CMYA5 may be involved in the same biological pathway and process. On the basis of this information, we performed replication studies for these three single-nucleotide polymorphisms. The rs3828611 was found to have conflicting results in our Irish samples and was dropped out without further investigation. The other two markers were verified in 23 other independent data sets. In a meta-analysis of all 23 replication samples (family samples, 912 families with 4160 subjects; case-control samples, 11 380 cases and 15 021 controls), we found that both markers are significantly associated with schizophrenia (rs10043986, odds ratio (OR)=1.11, 95% confidence interval (CI)=1.04-1.18, P=8.2 × 10(-4) and rs4704591, OR=1.07, 95% CI=1.03-1.11, P=3.0 × 10(-4)). The results were also significant for the 22 Caucasian replication samples (rs10043986, OR=1.11, 95% CI=1.03-1.17, P=0.0026 and rs4704591, OR=1.07, 95% CI=1.02-1.11, P=0.0015). Furthermore, haplotype conditioned analyses indicated that the association signals observed at these two markers are independent. On the basis of these results, we concluded that CMYA5 is associated with schizophrenia and further investigation of the gene is warranted.
Collapse
Affiliation(s)
- X Chen
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23298-0424, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
Collaborators
René S Kahn, Don H Linszen, Jim van Os, Durk Wiersma, Richard Bruggeman, Wiepke Cahn, Lieuwe de Haan, Lydia Krabbendam, Inez Myin-Germeys, Michael C O'Donovan, George K Kirov, Nick J Craddock, Peter A Holmans, Nigel M Williams, Lyudmila Georgieva, Ivan Nikolov, N Norton, H Williams, Draga Toncheva, Vihra Milanova, Michael J Owen, Christina M Hultman, Paul Lichtenstein, Emma F Thelander, Patrick Sullivan, Derek W Morris, Colm T O'Dushlaine, Elaine Kenny, Emma M Quinn, Michael Gill, Aiden Corvin, Andrew McQuillin, Khalid Choudhury, Susmita Datta, Jonathan Pimm, Srinivasa Thirumalai, Vinay Puri, Robert Krasucki, Jacob Lawrence, Digby Quested, Nicholas Bass, Hugh Gurling, Caroline Crombie, Gillian Fraser, Soh Leh Kuan, Nicholas Walker, David St Clair, Douglas H R Blackwood, Walter J Muir, Kevin A McGhee, Ben Pickard, Pat Malloy, Alan W Maclean, Margaret Van Beck, Naomi R Wray, Stuart Macgregor, Peter M Visscher, Michele T Pato, Helena Medeiros, Frank Middleton, Celia Carvalho, Christopher Morley, Ayman Fanous, David Conti, James A Knowles, Carlos Paz Ferreira, Antonio Macedo, M Helena Azevedo, Carlos N Pato, Jennifer L Stone, Douglas M Ruderfer, Andrew N Kirby, Manuel A R Ferreira, Mark J Daly, Shaun M Purcell, Pamela Sklar, Shaun M Purcell, Jennifer L Stone, Kimberly Chambert, Douglas M Ruderfer, Finny Kuruvilla, Stacey B Gabriel, Kristin Ardlie, Jennifer L Moran, Mark J Daly, Edward M Scolnick, Pamela Sklar,
Collapse
|
152
|
Rodriguez-Murillo L, Gogos JA, Karayiorgou M. The genetic architecture of schizophrenia: new mutations and emerging paradigms. Annu Rev Med 2011; 63:63-80. [PMID: 22034867 DOI: 10.1146/annurev-med-072010-091100] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although a genetic component of schizophrenia has been acknowledged for a long time, the underlying architecture of the genetic risk remains a contentious issue. Early linkage and candidate association studies led to largely inconclusive results. More recently, the availability of powerful technologies, samples of sufficient sizes, and genome-wide panels of genetic markers facilitated systematic and agnostic scans throughout the genome for either common or rare disease risk variants of small or large effect size, respectively. Although the former had limited success, the role of rare genetic events, such as copy-number variants (CNVs) or rare point mutations, has become increasingly important in gene discovery for schizophrenia. Importantly, recent research building upon earlier findings of de novo recurrent CNVs at the 22q11.2 locus, has highlighted a de novo mutational paradigm as a major component of the genetic architecture of schizophrenia. Recent progress is bringing us closer to earlier intervention and new therapeutic targets.
Collapse
|
153
|
Talkowski ME, Mullegama SV, Rosenfeld JA, van Bon BWM, Shen Y, Repnikova EA, Gastier-Foster J, Thrush DL, Kathiresan S, Ruderfer DM, Chiang C, Hanscom C, Ernst C, Lindgren AM, Morton CC, An Y, Astbury C, Brueton LA, Lichtenbelt KD, Ades LC, Fichera M, Romano C, Innis JW, Williams CA, Bartholomew D, Van Allen MI, Parikh A, Zhang L, Wu BL, Pyatt RE, Schwartz S, Shaffer LG, de Vries BBA, Gusella JF, Elsea SH. Assessment of 2q23.1 microdeletion syndrome implicates MBD5 as a single causal locus of intellectual disability, epilepsy, and autism spectrum disorder. Am J Hum Genet 2011; 89:551-63. [PMID: 21981781 DOI: 10.1016/j.ajhg.2011.09.011] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 09/17/2011] [Accepted: 09/19/2011] [Indexed: 12/15/2022] Open
Abstract
Persons with neurodevelopmental disorders or autism spectrum disorder (ASD) often harbor chromosomal microdeletions, yet the individual genetic contributors within these regions have not been systematically evaluated. We established a consortium of clinical diagnostic and research laboratories to accumulate a large cohort with genetic alterations of chromosomal region 2q23.1 and acquired 65 subjects with microdeletion or translocation. We sequenced translocation breakpoints; aligned microdeletions to determine the critical region; assessed effects on mRNA expression; and examined medical records, photos, and clinical evaluations. We identified a single gene, methyl-CpG-binding domain 5 (MBD5), as the only locus that defined the critical region. Partial or complete deletion of MBD5 was associated with haploinsufficiency of mRNA expression, intellectual disability, epilepsy, and autistic features. Fourteen alterations, including partial deletions of noncoding regions not typically captured or considered pathogenic by current diagnostic screening, disrupted MBD5 alone. Expression profiles and clinical characteristics were largely indistinguishable between MBD5-specific alteration and deletion of the entire 2q23.1 interval. No copy-number alterations of MBD5 were observed in 7878 controls, suggesting MBD5 alterations are highly penetrant. We surveyed MBD5 coding variations among 747 ASD subjects compared to 2043 non-ASD subjects analyzed by whole-exome sequencing and detected an association with a highly conserved methyl-CpG-binding domain missense variant, p.79Gly>Glu (c.236G>A) (p = 0.012). These results suggest that genetic alterations of MBD5 cause features of 2q23.1 microdeletion syndrome and that this epigenetic regulator significantly contributes to ASD risk, warranting further consideration in research and clinical diagnostic screening and highlighting the importance of chromatin remodeling in the etiology of these complex disorders.
Collapse
Affiliation(s)
- Michael E Talkowski
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Duncan LE, Keller MC. A critical review of the first 10 years of candidate gene-by-environment interaction research in psychiatry. Am J Psychiatry 2011; 168:1041-9. [PMID: 21890791 PMCID: PMC3222234 DOI: 10.1176/appi.ajp.2011.11020191] [Citation(s) in RCA: 679] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Gene-by-environment interaction (G×E) studies in psychiatry have typically been conducted using a candidate G×E (cG×E) approach, analogous to the candidate gene association approach used to test genetic main effects. Such cG×E research has received widespread attention and acclaim, yet cG×E findings remain controversial. The authors examined whether the many positive cG×E findings reported in the psychiatric literature were robust or if, in aggregate, cG×E findings were consistent with the existence of publication bias, low statistical power, and a high false discovery rate. METHOD The authors conducted analyses on data extracted from all published studies (103 studies) from the first decade (2000-2009) of cG×E research in psychiatry. RESULTS Ninety-six percent of novel cG×E studies were significant compared with 27% of replication attempts. These findings are consistent with the existence of publication bias among novel cG×E studies, making cG×E hypotheses appear more robust than they actually are. There also appears to be publication bias among replication attempts because positive replication attempts had smaller average sample sizes than negative ones. Power calculations using observed sample sizes suggest that cG×E studies are underpowered. Low power along with the likely low prior probability of a given cG×E hypothesis being true suggests that most or even all positive cG×E findings represent type I errors. CONCLUSIONS In this new era of big data and small effects, a recalibration of views about groundbreaking findings is necessary. Well-powered direct replications deserve more attention than novel cG×E findings and indirect replications.
Collapse
|
155
|
Gejman PV, Sanders AR, Kendler KS. Genetics of Schizophrenia: New Findings and Challenges. Annu Rev Genomics Hum Genet 2011; 12:121-44. [DOI: 10.1146/annurev-genom-082410-101459] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Pablo V. Gejman
- Center for Psychiatric Genetics, NorthShore University HealthSystem Research Institute, and University of Chicago, Evanston, Illinois 60201;
| | - Alan R. Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem Research Institute, and University of Chicago, Evanston, Illinois 60201;
| | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics and Departments of Psychiatry and Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298;
| |
Collapse
|
156
|
Lee KW, Woon PS, Teo YY, Sim K. Genome wide association studies (GWAS) and copy number variation (CNV) studies of the major psychoses: what have we learnt? Neurosci Biobehav Rev 2011; 36:556-71. [PMID: 21946175 DOI: 10.1016/j.neubiorev.2011.09.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 09/03/2011] [Accepted: 09/13/2011] [Indexed: 12/29/2022]
Abstract
Schizophrenia (SZ) and bipolar disorder (BPD) have high heritabilities and are clinically and genetically complex. Genome wide association studies (GWAS) and studies of copy number variations (CNV) in SZ and BPD have allowed probing of their underlying genetic risks. In this systematic review, we assess extant genetic signals from published GWAS and CNV studies of SZ and BPD up till March 2011. Risk genes associated with SZ at genome wide significance level (p value<7.2 × 10(-8)) include zinc finger binding protein 804A (ZNF804A), major histocompatibility (MHC) region on chromosome 6, neurogranin (NRGN) and transcription factor 4 (TCF4). Risk genes associated with BPD include ankyrin 3, node of Ranvier (ANK3), calcium channel, voltage dependent, L type, alpha 1C subunit (CACNA1C), diacylglycerol kinase eta (DGKH), gene locus on chromosome 16p12, and polybromo-1 (PBRM1) and very recently neurocan gene (NCAN). Possible common genes underlying psychosis include ZNF804A, CACNA1C, NRGN and PBRM1. The CNV studies suggest that whilst CNVs are found in both SZ and BPD, the large deletions and duplications are more likely found in SZ rather than BPD. The validation of any genetic signal is likely confounded by genetic and phenotypic heterogeneities which are influenced by epistatic, epigenetic and gene-environment interactions. There is a pressing need to better integrate the multiple research platforms including systems biology computational models, genomics, cross disorder phenotyping studies, transcriptomics, proteomics, metabolomics, neuroimaging and clinical correlations in order to get us closer to a more enlightened understanding of the genetic and biological basis underlying these potentially crippling conditions.
Collapse
Affiliation(s)
- Kok Wei Lee
- Institute of Mental Health/Woodbridge Hospital 10, Buangkok View, Singapore 539747, Singapore
| | | | | | | |
Collapse
|
157
|
Zhang K, Zhang L, Zhang W, Wang J. Pathway-based analysis for genome-wide association studies of schizophrenia to provide new insight in schizophrenia study. CHINESE SCIENCE BULLETIN-CHINESE 2011. [DOI: 10.1007/s11434-011-4742-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
158
|
Chen J, Lee G, Fanous AH, Zhao Z, Jia P, O'Neill A, Walsh D, Kendler KS, Chen X, The International Schizophrenia Consortium 1. Two non-synonymous markers in PTPN21, identified by genome-wide association study data-mining and replication, are associated with schizophrenia. Schizophr Res 2011; 131:43-51. [PMID: 21752600 PMCID: PMC4117700 DOI: 10.1016/j.schres.2011.06.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 06/15/2011] [Accepted: 06/20/2011] [Indexed: 01/27/2023]
Abstract
We conducted data-mining analyses of genome wide association (GWA) studies of the CATIE and MGS-GAIN datasets, and found 13 markers in the two physically linked genes, PTPN21 and EML5, showing nominally significant association with schizophrenia. Linkage disequilibrium (LD) analysis indicated that all 7 markers from PTPN21 shared high LD (r(2)>0.8), including rs2274736 and rs2401751, the two non-synonymous markers with the most significant association signals (rs2401751, P=1.10 × 10(-3) and rs2274736, P=1.21 × 10(-3)). In a meta-analysis of all 13 replication datasets with a total of 13,940 subjects, we found that the two non-synonymous markers are significantly associated with schizophrenia (rs2274736, OR=0.92, 95% CI: 0.86-0.97, P=5.45 × 10(-3) and rs2401751, OR=0.92, 95% CI: 0.86-0.97, P=5.29 × 10(-3)). One SNP (rs7147796) in EML5 is also significantly associated with the disease (OR=1.08, 95% CI: 1.02-1.14, P=6.43 × 10(-3)). These 3 markers remain significant after Bonferroni correction. Furthermore, haplotype conditioned analyses indicated that the association signals observed between rs2274736/rs2401751 and rs7147796 are statistically independent. Given the results that 2 non-synonymous markers in PTPN21 are associated with schizophrenia, further investigation of this locus is warranted.
Collapse
Affiliation(s)
- Jingchun Chen
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Suite 390, 800 E. Leigh Street, Richmond, VA 23298, USA,Department of Psychiatry, Virginia Commonwealth University, Suite 390A, 800 E. Leigh Street, Richmond, VA 23298, USA
| | - Grace Lee
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Suite 390, 800 E. Leigh Street, Richmond, VA 23298, USA,Department of Psychiatry, Virginia Commonwealth University, Suite 390A, 800 E. Leigh Street, Richmond, VA 23298, USA
| | - Ayman H. Fanous
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Suite 390, 800 E. Leigh Street, Richmond, VA 23298, USA,Department of Psychiatry, Virginia Commonwealth University, Suite 390A, 800 E. Leigh Street, Richmond, VA 23298, USA,Washington VA Medical Center, Washington, DC 20422, USA,Department of Psychiatry, Georgetown University School of Medicine, Washington, DC 20057, USA
| | - Zhongming Zhao
- Departments of Biomedical Informatics and Psychiatry, Vanderbilt University, 2525 West End Avenue, Nashville, TN 37203, USA
| | - Peilin Jia
- Departments of Biomedical Informatics and Psychiatry, Vanderbilt University, 2525 West End Avenue, Nashville, TN 37203, USA
| | - Anthony O'Neill
- The Department of Psychiatry, The Queens University, Belfast, Northern Ireland, UK
| | | | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Suite 390, 800 E. Leigh Street, Richmond, VA 23298, USA,Department of Psychiatry, Virginia Commonwealth University, Suite 390A, 800 E. Leigh Street, Richmond, VA 23298, USA,Department of Molecular and Human Genetics, Virginia Commonwealth University, Suite 390, 800 E. Leigh Street, Richmond, VA 23298, USA
| | - Xiangning Chen
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Suite 390, 800 E. Leigh Street, Richmond, VA 23298, USA,Department of Psychiatry, Virginia Commonwealth University, Suite 390A, 800 E. Leigh Street, Richmond, VA 23298, USA,Department of Molecular and Human Genetics, Virginia Commonwealth University, Suite 390, 800 E. Leigh Street, Richmond, VA 23298, USA,Corresponding author at: Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Suite 390A, 800 E. Leigh Street, Richmond, VA 23298, USA. Tel.: +1 804 828 8124; fax: +1 804 628 1035
| | | |
Collapse
|
159
|
Abstract
In genome-wide association studies, population stratification is recognized as producing inflated type I error due to the inflation of test statistics. Principal component-based methods applied to genotypes provide information about population structure, and have been widely used to control for stratification. Here we explore the precise relationship between genotype principal components and inflation of association test statistics, thereby drawing a connection between principal component-based stratification control and the alternative approach of genomic control. Our results provide an inherent justification for the use of principal components, but call into question the popular practice of selecting principal components based on significance of eigenvalues alone. We propose a new approach, called EigenCorr, which selects principal components based on both their eigenvalues and their correlation with the (disease) phenotype. Our approach tends to select fewer principal components for stratification control than does testing of eigenvalues alone, providing substantial computational savings and improvements in power. Analyses of simulated and real data demonstrate the usefulness of the proposed approach.
Collapse
Affiliation(s)
- Seunggeun Lee
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | | | |
Collapse
|
160
|
Flatscher-Bader T, Foldi CJ, Chong S, Whitelaw E, Moser RJ, Burne THJ, Eyles DW, McGrath JJ. Increased de novo copy number variants in the offspring of older males. Transl Psychiatry 2011; 1:e34. [PMID: 22832608 PMCID: PMC3309504 DOI: 10.1038/tp.2011.30] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 07/08/2011] [Indexed: 01/26/2023] Open
Abstract
The offspring of older fathers have an increased risk of neurodevelopmental disorders, such as schizophrenia and autism. In light of the evidence implicating copy number variants (CNVs) with schizophrenia and autism, we used a mouse model to explore the hypothesis that the offspring of older males have an increased risk of de novo CNVs. C57BL/6J sires that were 3- and 12-16-months old were mated with 3-month-old dams to create control offspring and offspring of old sires, respectively. Applying genome-wide microarray screening technology, 7 distinct CNVs were identified in a set of 12 offspring and their parents. Competitive quantitative PCR confirmed these CNVs in the original set and also established their frequency in an independent set of 77 offspring and their parents. On the basis of the combined samples, six de novo CNVs were detected in the offspring of older sires, whereas none were detected in the control group. Two of the CNVs were associated with behavioral and/or neuroanatomical phenotypic features. One of the de novo CNVs involved Auts2 (autism susceptibility candidate 2), and other CNVs included genes linked to schizophrenia, autism and brain development. This is the first experimental demonstration that the offspring of older males have an increased risk of de novo CNVs. Our results support the hypothesis that the offspring of older fathers have an increased risk of neurodevelopmental disorders such as schizophrenia and autism by generation of de novo CNVs in the male germline.
Collapse
Affiliation(s)
- T Flatscher-Bader
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
- The Queensland Institute of Medical Research, Herston, QLD, Australia
| | - C J Foldi
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - S Chong
- The Queensland Institute of Medical Research, Herston, QLD, Australia
| | - E Whitelaw
- The Queensland Institute of Medical Research, Herston, QLD, Australia
| | | | - T H J Burne
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia
| | - D W Eyles
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia
| | - J J McGrath
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia
- Discipline of Psychiatry, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
161
|
A genome-wide linkage scan for distinct subsets of schizophrenia characterized by age at onset and neurocognitive deficits. PLoS One 2011; 6:e24103. [PMID: 21897869 PMCID: PMC3163684 DOI: 10.1371/journal.pone.0024103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 07/30/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND As schizophrenia is genetically and phenotypically heterogeneous, targeting genetically informative phenotypes may help identify greater linkage signals. The aim of the study is to evaluate the genetic linkage evidence for schizophrenia in subsets of families with earlier age at onset or greater neurocognitive deficits. METHODS Patients with schizophrenia (n = 1,207) and their first-degree relatives (n = 1,035) from 557 families with schizophrenia were recruited from six data collection field research centers throughout Taiwan. Subjects completed a face-to-face semi-structured interview, the Continuous Performance Test (CPT), the Wisconsin Card Sorting Test, and were genotyped with 386 microsatellite markers across the genome. RESULTS A maximum nonparametric logarithm of odds (LOD) score of 4.17 at 2q22.1 was found in 295 families ranked by increasing age at onset, which had significant increases in the maximum LOD score compared with those obtained in initial linkage analyses using all available families. Based on this subset, a further subsetting by false alarm rate on the undegraded and degraded CPT obtained further increase in the nested subset-based LOD on 2q22.1, with a score of 7.36 in 228 families and 7.71 in 243 families, respectively. CONCLUSION We found possible evidence of linkage on chromosome 2q22.1 in families of schizophrenia patients with more CPT false alarm rates nested within the families with younger age at onset. These results highlight the importance of incorporating genetically informative phenotypes in unraveling the complex genetics of schizophrenia.
Collapse
|
162
|
Lee PH, Bergen SE, Perlis RH, Sullivan PF, Sklar P, Smoller JW, Purcell SM. Modifiers and subtype-specific analyses in whole-genome association studies: a likelihood framework. Hum Hered 2011; 72:10-20. [PMID: 21849790 DOI: 10.1159/000327158] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 03/07/2011] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE We propose new statistical methods for analyzing genetic case/control association data in which cases can be further classified into subtypes, for example, based on clinical features. The primary utility of our work is the ability to distinguish between subtype-specific and modifier effects of genetic variants within a single testing framework. METHODS A range of disease/subtype causal models are defined for genetic variants involving subtype-specific and modifier effects. We present a log-linear modeling framework enabling comparison between these causal models and selection of the best-fit model. RESULTS We evaluate and compare the analytic power and model selection performance of the proposed work with standard two-group-based association tests. Simulation studies demonstrate that our approach has similar or greater power than the traditional approach over a range of causal models. We also report empirical findings about the impact of misspecification of subtype frequency during model selection, and extend the application of the proposed work to the cross-disorder association studies of multiple diseases. CONCLUSION Whether a variant is a disease risk factor, is subtype specific, or modifies disease features has important consequences for the interpretation and follow-up of genetic associations. Our framework provides a simple, systematic way to evaluate and describe associations involving such subtype-specific or modifier effects.
Collapse
Affiliation(s)
- Phil H Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Department of Psychiatry, Harvard Medical School, Boston, Mass., USA
| | | | | | | | | | | | | |
Collapse
|
163
|
Support for association of HSPG2 with tardive dyskinesia in Caucasian populations. THE PHARMACOGENOMICS JOURNAL 2011; 12:513-20. [PMID: 21808285 DOI: 10.1038/tpj.2011.32] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tardive dyskinesia (TD) is a severe adverse effect of chronic antipsychotic drug treatment. In addition to clinical risk factors, TD susceptibility is influenced by genetic predisposition. Recently, Syu et al. (2010) reported a genome-wide association screening of TD in Japanese schizophrenia patients. The best result was association of single-nucleotide polymorphism (SNP) rs2445142 in the HSPG2 (heparan sulfate proteoglycan 2) gene with TD. In the present study, we report a replication study of the five top Japanese TD-associated SNPs in two Caucasian TD samples. Applying logistic regression and controlling for relevant clinical risk factors, we were able to replicate the association of HSPG2 SNP rs2445142 with TD in a prospective study sample of 179 Americans of European origin by performing a secondary analysis of the CATIE (Clinical Antipsychotic Trials of Intervention Effectiveness) genome-wide association study data set, and using a perfect proxy surrogate marker (rs878949; P = 0.039). An association of the 'G' risk allele of HSPG2 SNP rs2445142 with TD was also shown in a sample of Jewish Israeli schizophrenia patients (retrospective, cross-sectional design; P = 0.03). Although the associations were only nominally significant, the findings provide further support for the possible involvement of HSPG2 in susceptibility to TD.
Collapse
|
164
|
Alkelai A, Lupoli S, Greenbaum L, Giegling I, Kohn Y, Sarner-Kanyas K, Ben-Asher E, Lancet D, Rujescu D, Macciardi F, Lerer B. Identification of new schizophrenia susceptibility loci in an ethnically homogeneous, family-based, Arab-Israeli sample. FASEB J 2011; 25:4011-23. [PMID: 21795503 DOI: 10.1096/fj.11-184937] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
While the use of population-based samples is a common strategy in genome-wide association studies (GWASs), family-based samples have considerable advantages, such as robustness against population stratification and false-positive associations, better quality control, and the possibility to check for both linkage and association. In a genome-wide linkage study of schizophrenia in Arab-Israeli families with multiple affected individuals, we previously reported significant evidence for a susceptibility locus at chromosome 6q23.2-q24.1 and suggestive evidence at chromosomes 10q22.3-26.3, 2q36.1-37.3 and 7p21.1-22.3. To identify schizophrenia susceptibility genes, we applied a family-based GWAS strategy in an enlarged, ethnically homogeneous, Arab-Israeli family sample. We performed genome-wide single nucleotide polymorphism (SNP) genotyping and single SNP transmission disequilibrium test association analysis and found genome-wide significant association (best value of P=1.22×10(-11)) for 8 SNPs within or near highly reasonable functional candidate genes for schizophrenia. Of particular interest are a group of SNPs within and flanking the transcriptional factor LRRFIP1 gene. To determine replicability of the significant associations beyond the Arab-Israeli population, we studied the association of the significant SNPs in a German case-control validation sample and found replication of associations near the UGT1 subfamily and EFHD1 genes. Applying an exploratory homozygosity mapping approach as a complementary strategy to identify schizophrenia susceptibility genes in our Arab Israeli sample, we identified 8 putative disease loci. Overall, this GWAS, which emphasizes the important contribution of family based studies, identifies promising candidate genes for schizophrenia.
Collapse
Affiliation(s)
- Anna Alkelai
- Biological Psychiatry Laboratory, Department of Psychiatry, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Bigos KL, Bies RR, Pollock BG, Lowy JJ, Zhang F, Weinberger DR. Genetic variation in CYP3A43 explains racial difference in olanzapine clearance. Mol Psychiatry 2011; 16:620-5. [PMID: 21519338 PMCID: PMC3100476 DOI: 10.1038/mp.2011.38] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 02/15/2011] [Indexed: 12/24/2022]
Abstract
The antipsychotic drug, olanzapine, one of the most widely used drugs in clinical medicine, has a high rate of discontinuation due to inefficacy and/or adverse effects. We identified a single nucleotide polymorphism in the drug metabolizing enzyme, cytochrome P450 3A43 (CYP3A43; rs472660), that highly significantly predicted olanzapine clearance in the Clinical Antipsychotic Trials of Intervention Effectiveness trial (P=5.9e(-7)). Moreover, at standard antipsychotic doses, 50% of individuals with the high clearance genotype (AA) have trough blood levels below the therapeutic range. Interestingly, a much higher proportion of African Americans carry the A allele compared with Caucasians (allele frequency 67 vs 14%). After accounting for CYP3A43 genotype, race is no longer a significant predictor of olanzapine clearance. Olanzapine clearance was associated with measures of clinical response. Patients with greater clearance had higher symptom ratings and were more likely to discontinue treatment due to an inadequate response. Our data identify a genetic mechanism for variation in olanzapine response and demonstrate that blood level monitoring of olanzapine treatment is advisable.
Collapse
Affiliation(s)
- Kristin L. Bigos
- Genes, Cognition, and Psychosis Program, National Institute of Mental Health, NIH, Bethesda, MD
| | - Robert R. Bies
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Bruce G. Pollock
- Rotman Research Institute, University of Toronto, Toronto, Ontario, Canada
- Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | - Joshua J. Lowy
- Genes, Cognition, and Psychosis Program, National Institute of Mental Health, NIH, Bethesda, MD
| | - Fengyu Zhang
- Genes, Cognition, and Psychosis Program, National Institute of Mental Health, NIH, Bethesda, MD
| | - Daniel R. Weinberger
- Genes, Cognition, and Psychosis Program, National Institute of Mental Health, NIH, Bethesda, MD
- Clinical Brain Disorders Branch, National Institute of Mental Health, NIH, Bethesda, MD
| |
Collapse
|
166
|
Zhang F, Chen Q, Ye T, Lipska BK, Straub RE, Vakkalanka R, Rujescu D, St Clair D, Hyde TM, Bigelow L, Kleinman JE, Weinberger DR. Evidence of sex-modulated association of ZNF804A with schizophrenia. Biol Psychiatry 2011; 69:914-7. [PMID: 21349497 DOI: 10.1016/j.biopsych.2011.01.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 11/29/2010] [Accepted: 01/03/2011] [Indexed: 12/14/2022]
Abstract
BACKGROUND The single nucleotide polymorphism (SNP) rs1344706 in ZNF804A (2q32.1) has been associated with schizophrenia in a genome-wide association study (GWAS). A recent candidate gene study, which replicated the positive association with rs1344706, identified another positive SNP (rs7597593) in ZNF804A associated with schizophrenia. METHODS We performed an association study of rs7597593 in four GWAS cohorts of European ancestry. Postmortem human brain expression data of normal Caucasian individuals (n = 89) was also analyzed for examining the effect of rs7597593 on ZNF804A messenger RNA expression, using logistic regression and linear regression. RESULTS We found that rs7597593 was significantly associated with schizophrenia in the combined GWAS datasets (n = 5023, odds ratio [OR](combined) = 1.15, p = .0011). Analysis of stratification by sex showed that the association was driven by the female subjects (OR = 1.29, p = .0002) and was not significant in male subjects (OR = 1.08, p = .148) in the combined sample of four cohorts. A sex by genotype interaction was near significant in both the Genetic Association Information Network sample (p = .0532) and the combined sample of four cohorts (p(combined) = .0531). Gene expression analysis showed no main effects but a significant female-specific association (p(female) = .047, p(male) = .335) and sex by genotype interaction (p = .0166) for rs7597593. CONCLUSIONS Our data suggest a clinical and molecular modulation by sex of the association of ZNF804A SNP rs7597593 and risk of schizophrenia.
Collapse
Affiliation(s)
- Fengyu Zhang
- Genes, Cognition and Psychosis Program and Clinical, Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Abstract
The purpose of this invited review is to summarize the state of genetic research into the etiology of schizophrenia (SCZ) and to consider options for progress. The fundamental uncertainty in SCZ genetics has always been the nature of the beast, the underlying genetic architecture. If this were known, studies using the appropriate technologies and sample sizes could be designed with an excellent chance of producing high-confidence results. Until recently, few pertinent data were available, and the field necessarily relied on speculation. However, for the first time in the complex and frustrating history of inquiry into the genetics of SCZ, we now have empirical data about the genetic basis of SCZ that implicate specific loci and that can be used to plan the next steps forward.
Collapse
Affiliation(s)
- Yunjung Kim
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Stephanie Zerwas
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC
| | - Sara E. Trace
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina, Chapel Hill, NC
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
168
|
Sim K, Lee J, Subramaniam M, Liu JJ, Keefe R, Zhang XD, Lee TS, Chong SA. Integrated genetic and genomic approach in the SingaporeTranslational and Clinical Research in Psychosis Study: an overview. Early Interv Psychiatry 2011; 5:91-9. [PMID: 21535421 DOI: 10.1111/j.1751-7893.2011.00272.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AIMS Schizophrenia is a severe mental disorder with onset frequently in adolescence and followed by a chronic and disabling course. Although the exact pathophysiology of this devastating disorder has not been clearly elucidated, a large part of it has been attributed to genetic influences. This article seeks to provide an overview on what our group has embarked on--to elucidate genetic risk factors for schizophrenia within the Chinese ethnic group. METHODS We plan to conduct an integrated approach to interrogate comprehensively the genome from different angles and in stages. The first stage involves a genome-wide association study of 1000 cases of schizophrenia-control pairs, with a follow-up replication study in another 1000 cases of schizophrenia and in 1000 controls, and combination analyses with groups from other places including China and Hong Kong. Other than the genome-wide association study, gene sequencing for purported candidate genes and copy number variation analysis will be performed. Neurocognitive intermediate phenotypes will be employed to deconstruct the complex schizophrenia phenotype in a bid to improve association findings. Promising leads from longitudinal gene and protein expression in ultra-high-risk subjects who develop psychosis and schizophrenia (in a parallel study) will be followed up as candidate genes and sequenced in the genetic analysis. Functional analysis forms the last stage of this integrated approach. CONCLUSION This integrated genetic and genomic approach will hopefully help in further characterizing and deepening our understanding of molecular pathophysiological mechanisms underlying schizophrenia.
Collapse
Affiliation(s)
- Kang Sim
- Department of General Psychiatry, Woodbridge Hospital/Institute of Mental Health Research Division, Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
169
|
Abstract
Pathway-based analysis is rapidly emerging as an alternative but powerful approach for searching for disease causal genes from genomic datasets and has been applied to many complex diseases recently, but it is only now beginning to be applied in psychiatry. Here, we discuss critical issues in the pathway-based approach by specifically comparing the first pathway analysis of genome-wide association studies (GWAS) datasets in neuropsychiatric disorders by O'Dushlaine and colleagues (Molecular Psychiatry 2010, doi:10.1038/mp.2010.7) with our analysis. We also computed the power of gene set enrichment analysis, hypergeometric test, and SNP ratio test in order to assist future applications of these methods in pathway-based analysis of GWAS datasets. Overall, we suggest that the pathway-based approach is effective but caution is needed in interpreting the results of such analysis.
Collapse
|
170
|
Tang B, Thornton-Wells T, Askland KD. Comparative linkage meta-analysis reveals regionally-distinct, disparate genetic architectures: application to bipolar disorder and schizophrenia. PLoS One 2011; 6:e19073. [PMID: 21559500 PMCID: PMC3084739 DOI: 10.1371/journal.pone.0019073] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Accepted: 03/25/2011] [Indexed: 11/18/2022] Open
Abstract
New high-throughput, population-based methods and next-generation sequencing capabilities hold great promise in the quest for common and rare variant discovery and in the search for "missing heritability." However, the optimal analytic strategies for approaching such data are still actively debated, representing the latest rate-limiting step in genetic progress. Since it is likely a majority of common variants of modest effect have been identified through the application of tagSNP-based microarray platforms (i.e., GWAS), alternative approaches robust to detection of low-frequency (1-5% MAF) and rare (<1%) variants are of great importance. Of direct relevance, we have available an accumulated wealth of linkage data collected through traditional genetic methods over several decades, the full value of which has not been exhausted. To that end, we compare results from two different linkage meta-analysis methods--GSMA and MSP--applied to the same set of 13 bipolar disorder and 16 schizophrenia GWLS datasets. Interestingly, we find that the two methods implicate distinct, largely non-overlapping, genomic regions. Furthermore, based on the statistical methods themselves and our contextualization of these results within the larger genetic literatures, our findings suggest, for each disorder, distinct genetic architectures may reside within disparate genomic regions. Thus, comparative linkage meta-analysis (CLMA) may be used to optimize low-frequency and rare variant discovery in the modern genomic era.
Collapse
Affiliation(s)
- Brady Tang
- Biostatistics Graduate Program, Brown University, Providence, Rhode Island, United States of America
| | - Tricia Thornton-Wells
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Kathleen D. Askland
- Department of Psychiatry and Human Behavior, Butler Hospital, The Warren Alpert School of Medicine of Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
171
|
Ramsey TL, Meltzer HY, Brock GN, Mehrotra B, Jayathilake K, Bobo WV, Brennan MD. Evidence for a SULT4A1 haplotype correlating with baseline psychopathology and atypical antipsychotic response. Pharmacogenomics 2011; 12:471-80. [PMID: 21521020 PMCID: PMC3168511 DOI: 10.2217/pgs.10.205] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM This study evaluated the impact of SULT4A1 gene variation on psychopathology and antipsychotic drug response in Caucasian subjects from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) study and a replication sample. PATIENTS & METHODS SULT4A1 haplotypes were determined using SNP data. The relationship to baseline psychopathology was evaluated using linear regression of Positive and Negative Syndrome Scale (PANSS) total score. Drug response was evaluated using Mixed Model Repeat Measures (MMRM) for change in PANSS. RESULTS For the CATIE sample, patients carrying a haplotype designated SULT4A1-1(+) displayed higher baseline PANSS (p = 0.03) and, when treated with olanzapine, demonstrated a significant interaction with time (p = 0.009) in the MMRM. SULT4A1-1(+) patients treated with olanzapine displayed improved response compared with SULT4A1-1(-) patients treated with olanzapine (p = 0.008) or to SULT4A1-1(+) patients treated with risperidone (p = 0.006). In the replication sample, SULT4A1-1(+) patients treated with olanzapine demonstrated greater improvement than SULT4A1-1(-) patients treated with olanzapine (p = 0.05) or than SULT4A1-1(+) patients treated with risperidone (p = 0.05). CONCLUSION If validated, determination of SULT4A1-1 haplotype status might be useful for identifying patients who show an enhanced response to long-term olanzapine treatment. Original submitted 6 October 2010; Revision submitted 9 December 2010.
Collapse
Affiliation(s)
| | | | - Guy N Brock
- Department of Bioinformatics & Biostatistics, School of Public Health & Information Sciences, University of Louisville, Louisville, KY, USA
| | | | - Karu Jayathilake
- Psychiatric Hospital at Vanderbilt University, Nashville, TN, USA
| | - William V Bobo
- Psychiatric Hospital at Vanderbilt University, Nashville, TN, USA
| | - Mark D Brennan
- SureGene, LLC, 600 Envoy Circle, Louisville, KY 40299, USA
| |
Collapse
|
172
|
Williams HJ, Norton N, Dwyer S, Moskvina V, Nikolov I, Carroll L, Georgieva L, Williams NM, Morris DW, Quinn EM, Giegling I, Ikeda M, Wood J, Lencz T, Hultman C, Lichtenstein P, Thiselton D, Maher BS, Molecular Genetics of Schizophrenia Collaboration (MGS), International Schizophrenia Consortium (ISC), SGENE-plus, GROUP, Malhotra AK, Riley B, Kendler KS, Gill M, Sullivan P, Sklar P, Purcell S, Nimgaonkar VL, Kirov G, Holmans P, Corvin A, Rujescu D, Craddock N, Owen MJ, O’Donovan MC. Fine mapping of ZNF804A and genome-wide significant evidence for its involvement in schizophrenia and bipolar disorder. Mol Psychiatry 2011; 16:429-41. [PMID: 20368704 PMCID: PMC3918934 DOI: 10.1038/mp.2010.36] [Citation(s) in RCA: 214] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 02/15/2010] [Accepted: 02/16/2010] [Indexed: 12/16/2022]
Abstract
A recent genome-wide association study (GWAS) reported evidence for association between rs1344706 within ZNF804A (encoding zinc-finger protein 804A) and schizophrenia (P=1.61 × 10(-7)), and stronger evidence when the phenotype was broadened to include bipolar disorder (P=9.96 × 10(-9)). In this study we provide additional evidence for association through meta-analysis of a larger data set (schizophrenia/schizoaffective disorder N=18 945, schizophrenia plus bipolar disorder N=21 274 and controls N=38 675). We also sought to better localize the association signal using a combination of de novo polymorphism discovery in exons, pooled de novo polymorphism discovery spanning the genomic sequence of the locus and high-density linkage disequilibrium (LD) mapping. The meta-analysis provided evidence for association between rs1344706 that surpasses widely accepted benchmarks of significance by several orders of magnitude for both schizophrenia (P=2.5 × 10(-11), odds ratio (OR) 1.10, 95% confidence interval 1.07-1.14) and schizophrenia and bipolar disorder combined (P=4.1 × 10(-13), OR 1.11, 95% confidence interval 1.07-1.14). After de novo polymorphism discovery and detailed association analysis, rs1344706 remained the most strongly associated marker in the gene. The allelic association at the ZNF804A locus is now one of the most compelling in schizophrenia to date, and supports the accumulating data suggesting overlapping genetic risk between schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Hywel J Williams
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Nadine Norton
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sarah Dwyer
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Valentina Moskvina
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Ivan Nikolov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Liam Carroll
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Lyudmila Georgieva
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Nigel M Williams
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Derek W Morris
- Neuropsychiatric Genetics Research Group, School of Medicine, Trinity College Dublin, Ireland
| | - Emma M Quinn
- Neuropsychiatric Genetics Research Group, School of Medicine, Trinity College Dublin, Ireland
| | - Ina Giegling
- Division of Molecular and Clinical Neurobiology, Ludwig-Maximilians-University, Nußbaumstr 7, 80336 Munich, Germany
| | - Masashi Ikeda
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
- Department of Psychiatry, Fujita Health University School of Medicine, Aichi 470-1192, Japan
| | - Joel Wood
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Todd Lencz
- Center for Translational Psychiatry, The Feinstein Institute for Medical Research, North Shore - Long Island Jewish Health System, Manhasset, NY USA
- Division of Research, The Zucker Hillside Hospital, Glen Oaks, NY USA
- Dept of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY USA
| | - Christina Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Paul Lichtenstein
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Dawn Thiselton
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Brion S Maher
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | | | | | - SGENE-plus
- Full author details and affiliations are given in acknowledgement section
| | - GROUP
- Full author details and affiliations are given in acknowledgement section
| | - Anil K Malhotra
- Center for Translational Psychiatry, The Feinstein Institute for Medical Research, North Shore - Long Island Jewish Health System, Manhasset, NY USA
- Division of Research, The Zucker Hillside Hospital, Glen Oaks, NY USA
- Dept of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY USA
| | - Brien Riley
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Human Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Kenneth S Kendler
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Human Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael Gill
- Neuropsychiatric Genetics Research Group, School of Medicine, Trinity College Dublin, Ireland
| | - Patrick Sullivan
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Pamela Sklar
- Psychiatric and Neurodevelopmental Genetics Unit
- Center for Human Genetic Research, Massachusetts General Hospital, Massachusetts 02114, USA
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- The Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Shaun Purcell
- Psychiatric and Neurodevelopmental Genetics Unit
- Center for Human Genetic Research, Massachusetts General Hospital, Massachusetts 02114, USA
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- The Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Vishwajit L Nimgaonkar
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - George Kirov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Peter Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, School of Medicine, Trinity College Dublin, Ireland
| | - Dan Rujescu
- Division of Molecular and Clinical Neurobiology, Ludwig-Maximilians-University, Nußbaumstr 7, 80336 Munich, Germany
| | - Nicholas Craddock
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Michael C O’Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
173
|
Saetre P, Vares M, Werge T, Andreassen OA, Arinami T, Ishiguro H, Nanko S, Tan EC, Han DH, Roffman JL, Muntjewerff JW, Jagodzinski PP, Kempisty B, Hauser J, Vilella E, Betcheva E, Nakamura Y, Regland B, Agartz I, Hall H, Terenius L, Jönsson EG. Methylenetetrahydrofolate reductase (MTHFR) C677T and A1298C polymorphisms and age of onset in schizophrenia: a combined analysis of independent samples. Am J Med Genet B Neuropsychiatr Genet 2011; 156:215-24. [PMID: 21302350 DOI: 10.1002/ajmg.b.31160] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 11/30/2010] [Indexed: 12/20/2022]
Abstract
Methylenetetrahydrofolate reductase (MTHFR) is involved in the one-carbon cycle, which is of importance for nucleotide synthesis and methylation of DNA, membranes, proteins and lipids. The MTHFR gene includes two common polymorphisms (rs1801133 or C677T; rs1801131 or A1298C) which both alter enzyme activity. The T-allele of the C677T polymorphism has recently been associated with earlier age at onset of schizophrenia. In the present study we examined the association between the MTHFR C677T and A1298C polymorphisms and age at onset of schizophrenia in twelve samples consisting of 3,213 unrelated schizophrenia patients, including the original Scandinavian sample. There was no consistent relationship between MTHFR C677T, A1298C or combined 677T/1298C carriers and age of onset in schizophrenia when the results of each study were combined using meta-analysis. The present results suggest that the investigated MTHFR polymorphisms do not influence age of onset in schizophrenia.
Collapse
Affiliation(s)
- Peter Saetre
- Department of Clinical Neuroscience, HUBIN Project, Karolinska Institutet and Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Schanze D, Ekici AB, Gawlik M, Pfuhlmann B, Reis A, Stöber G. Evaluation of risk loci for schizophrenia derived from genome-wide association studies in a German population. Am J Med Genet B Neuropsychiatr Genet 2011; 156:198-203. [PMID: 21302348 DOI: 10.1002/ajmg.b.31156] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 11/24/2010] [Indexed: 11/10/2022]
Abstract
In the genome-wide association study (GWAS) on schizophrenia [O'Donovan et al. (2008); Nat Genet 40:1053–1055] a UK-sample of 479 cases with DSM-IV schizophrenia was genotyped in comparison to control subjects with follow up of 12 putative loci in international replication sets of approximately 15,000 cases and controls. In these cohorts and a combined bipolar and schizophrenia UK-sample, six single nucleotide polymorphisms (SNPs) supported association, with the strongest evidence for SNP-marker rs1344706 at the zinc finger ZNF804A locus on chromosome 2q32.1 (P = 1.61 × 10−7). We attempted replication of these findings in a German population of 2,154 individuals (632 with affective disorders, 937 with schizophrenia, and 585 controls), but found none of the GWAS risk alleles significantly associated with psychosis. Particularly rs1344706, initially surpassing the genome-wide significance level in an extended phenotype of schizophrenia and affective disorder, produced consistently negative results. At the ZNF804A locus estimated Odds ratios reached 1.08 (0.93–1.26 95% CI) for the schizophrenia sample and 1.04 (0.90–1.20 95% CI) for the combined set of cases with schizophrenia and affective disorder. The main limitation of our study may be the reduced power of the sample size, but our data may be useful for future meta-analysis of GWA data sets. Although GWAS have proven extraordinary successful in identifying susceptibility genes for complex genetic disorders, the hypothesis of common genetic variants in the complex group of the schizophrenic psychoses with small effect size but relatively high frequency is still put to further scrutiny.
Collapse
Affiliation(s)
- D Schanze
- Institute of Human Genetics, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
175
|
Adkins DE, Aberg K, McClay JL, Bukszár J, Zhao Z, Jia P, Stroup TS, Perkins D, McEvoy JP, Lieberman JA, Sullivan PF, van den Oord EJCG. Genomewide pharmacogenomic study of metabolic side effects to antipsychotic drugs. Mol Psychiatry 2011; 16:321-32. [PMID: 20195266 PMCID: PMC2891163 DOI: 10.1038/mp.2010.14] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 12/09/2009] [Accepted: 01/20/2010] [Indexed: 12/24/2022]
Abstract
Understanding individual differences in the susceptibility to metabolic side effects as a response to antipsychotic therapy is essential to optimize the treatment of schizophrenia. Here, we perform genomewide association studies (GWAS) to search for genetic variation affecting the susceptibility to metabolic side effects. The analysis sample consisted of 738 schizophrenia patients, successfully genotyped for 492K single nucleotide polymorphisms (SNPs), from the genomic subsample of the Clinical Antipsychotic Trial of Intervention Effectiveness study. Outcomes included 12 indicators of metabolic side effects, quantifying antipsychotic-induced change in weight, blood lipids, glucose and hemoglobin A1c, blood pressure and heart rate. Our criterion for genomewide significance was a pre-specified threshold that ensures, on average, only 10% of the significant findings are false discoveries. A total of 21 SNPs satisfied this criterion. The top finding indicated that a SNP in Meis homeobox 2 (MEIS2) mediated the effects of risperidone on hip circumference (q=0.004). The same SNP was also found to mediate risperidone's effect on waist circumference (q=0.055). Genomewide significant finding were also found for SNPs in PRKAR2B, GPR98, FHOD3, RNF144A, ASTN2, SOX5 and ATF7IP2, as well as in several intergenic markers. PRKAR2B and MEIS2 both have previous research indicating metabolic involvement, and PRKAR2B has previously been shown to mediate antipsychotic response. Although our findings require replication and functional validation, this study shows the potential of GWAS to discover genes and pathways that potentially mediate adverse effects of antipsychotic medication.
Collapse
Affiliation(s)
- D E Adkins
- Center for Biomarker Research and Personalized Medicine, School of Pharmacy, Medical College of Virginia of Virginia Commonwealth University, Richmond, VA 23223, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Donohoe G, Rose E, Frodl T, Morris D, Spoletini I, Adriano F, Bernardini S, Caltagirone C, Bossù P, Gill M, Corvin AP, Spalletta G. ZNF804A risk allele is associated with relatively intact gray matter volume in patients with schizophrenia. Neuroimage 2011; 54:2132-7. [DOI: 10.1016/j.neuroimage.2010.09.089] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 09/09/2010] [Accepted: 09/27/2010] [Indexed: 11/16/2022] Open
|
177
|
McClay JL, Adkins DE, Aberg K, Bukszár J, Khachane AN, Keefe RSE, Perkins DO, McEvoy JP, Stroup TS, Vann RE, Beardsley PM, Lieberman JA, Sullivan PF, van den Oord EJCG. Genome-wide pharmacogenomic study of neurocognition as an indicator of antipsychotic treatment response in schizophrenia. Neuropsychopharmacology 2011; 36:616-26. [PMID: 21107309 PMCID: PMC3055694 DOI: 10.1038/npp.2010.193] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Revised: 09/16/2010] [Accepted: 09/23/2010] [Indexed: 01/16/2023]
Abstract
Neurocognitive deficits are a core feature of schizophrenia and, therefore, represent potentially critical outcome variables for assessing antipsychotic treatment response. We performed genome-wide association studies (GWAS) with 492K single nucleotide polymorphisms (SNPs) in a sample of 738 patients with schizophrenia from the Clinical Antipsychotic Trials of Intervention Effectiveness study. Outcome variables consisted of a neurocognitive battery administered at multiple time points over an 18-month period, measuring processing speed, verbal memory, vigilance, reasoning, and working memory domains. Genetic mediation of improvements in each of these five domains plus a composite neurocognitive measure was assessed for each of five antipsychotics (olanzapine, perphenazine, quetiapine, risperidone, and ziprasidone). Six SNPs achieved genome-wide significance using a pre-specified threshold that ensures, on average, only 1 in 10 findings is a false discovery. These six SNPs were located within, or in close proximity to, genes EHF, SLC26A9, DRD2, GPR137B, CHST8, and IL1A. The more robust findings, that is those significant across multiple neurocognitive domains and having adjacent SNPs showing evidence for association, were rs286913 at the EHF gene (p-value 6.99 × 10(-8), q-value 0.034, mediating the effects of ziprasidone on vigilance), rs11240594 at SLC26A9 (p-value 1.4 × 10(-7), q-value 0.068, mediating the effects of olanzapine on processing speed), and rs11677416 at IL1A (p-value 6.67 × 10(-7), q-value 0.081, mediating the effects of olanzapine on working memory). This study has generated several novel candidate genes for antipsychotic response. However, our findings will require replication and functional validation. To facilitate replication efforts, we provide all GWAS p-values for download.
Collapse
Affiliation(s)
- Joseph L McClay
- Center for Biomarker Research and Personalized Medicine, School of Pharmacy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0533, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Sun J, Wan C, Jia P, Fanous AH, Kendler KS, Riley BP, Zhao Z. Application of systems biology approach identifies and validates GRB2 as a risk gene for schizophrenia in the Irish Case Control Study of Schizophrenia (ICCSS) sample. Schizophr Res 2011; 125:201-8. [PMID: 21195589 PMCID: PMC3031722 DOI: 10.1016/j.schres.2010.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2010] [Revised: 12/02/2010] [Accepted: 12/06/2010] [Indexed: 02/02/2023]
Abstract
Recently, we prioritized 160 schizophrenia candidate genes (SZGenes) by integrating multiple lines of evidence and subsequently identified twenty-four pathways in which these 160 genes are overrepresented. Among them, four neurotransmitter-related pathways were top ranked. In this study, we extended our previous pathway analysis by applying a systems biology approach to identifying candidate genes for schizophrenia. We constructed protein-protein interaction subnetworks for four neurotransmitter-related pathways and merged them to obtain a general neurotransmitter network, from which five candidate genes stood out. We tested the association of four genes (GRB2, HSPA5, YWHAG, and YWHAZ) in the Irish Case-Control Study of Schizophrenia (ICCSS) sample (1021 cases and 626 controls). Interestingly, six of the seven tested SNPs in GRB2 showed significant signal, two of which (rs7207618 and rs9912608) remained significant after permutation test or Bonferroni correction, suggesting that GRB2 might be a risk gene for schizophrenia in Irish population. To our knowledge, this is the first report of GRB2 being significantly associated with schizophrenia in a specific population. Our results suggest that the systems biology approach is promising for identification of candidate genes and understanding the etiology of complex diseases.
Collapse
Affiliation(s)
- Jingchun Sun
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Chunling Wan
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Peilin Jia
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Ayman H. Fanous
- Washington VA Medical Center, Washington, DC 20422, USA
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Psychiatry, Georgetown University School of Medicine, Washington, DC 20007, USA
- Department of Psychiatry, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Brien P. Riley
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
179
|
Where are the missing pieces of the schizophrenia genetics puzzle? Curr Opin Genet Dev 2011; 21:310-6. [PMID: 21277191 DOI: 10.1016/j.gde.2011.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 12/17/2010] [Accepted: 01/03/2011] [Indexed: 12/15/2022]
Abstract
On the basis of recent data from candidate region/gene and genome-wide association studies (GWAS) and their follow-up investigations, the number of genes potentially implicated in schizophrenia has been estimated to be over 1000. However, with regard to the identified odds ratio, it is likely that genetic variants with more definitive effect on schizophrenia phenotype are still missing. The hunt therefore remains open for the genetic variants that would explain the majority of the missing heritability of schizophrenia. This review aims at summarizing data from recent DNA microarray and target gene/region resequencing in order to propose new insights of where to look next. The review is divided into three sections: GWAS, copy-number variations and rare variant--candidate gene resequencing.
Collapse
|
180
|
McClay JL, Adkins DE, Aberg K, Stroup S, Perkins DO, Vladimirov VI, Lieberman JA, Sullivan PF, van den Oord EJCG. Genome-wide pharmacogenomic analysis of response to treatment with antipsychotics. Mol Psychiatry 2011; 16:76-85. [PMID: 19721433 PMCID: PMC2888895 DOI: 10.1038/mp.2009.89] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 05/29/2009] [Accepted: 07/27/2009] [Indexed: 12/25/2022]
Abstract
Schizophrenia is an often devastating neuropsychiatric illness. Understanding the genetic variation affecting response to antipsychotics is important to develop novel diagnostic tests to match individual schizophrenia patients to the most effective and safe medication. In this study, we use a genome-wide approach to detect genetic variation underlying individual differences in response to treatment with the antipsychotics olanzapine, quetiapine, risperidone, ziprasidone and perphenazine. Our sample consisted of 738 subjects with DSM-IV schizophrenia who took part in the Clinical Antipsychotic Trials of Intervention Effectiveness. Subjects were genotyped using the Affymetrix 500 K genotyping platform plus a custom 164 K chip to improve genome-wide coverage. Treatment outcome was measured using the Positive and Negative Syndrome Scale. Our criterion for genome-wide significance was a prespecified threshold that ensures that, on an average, only 10% of the significant findings are false discoveries. The top statistical result reached significance at our prespecified threshold and involved a single-nucleotide polymorphism (SNP) in an intergenic region on chromosome 4p15. In addition, SNPs in Ankyrin Repeat and Sterile Alpha Motif Domain-Containing Protein 1B (ANKS1B) and in the Contactin-Associated Protein-Like 5 gene (CNTNAP5), which mediated the effects of olanzapine and risperidone on Negative symptoms, were very close to our threshold for declaring significance. The most significant SNP in CNTNAP5 is nonsynonymous, giving rise to an amino-acid substitution. In addition to highlighting our top results, we provide all P-values for download as a resource for investigators with the requisite samples to carry out replication. This study demonstrates the potential of genome-wide association studies to discover novel genes that mediate the effects of antipsychotics, which could eventually help to tailor drug treatment to schizophrenic patients.
Collapse
Affiliation(s)
- J L McClay
- Center for Biomarker Research and Personalized Medicine, School of Pharmacy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Kurian SM, Le-Niculescu H, Patel SD, Bertram D, Davis J, Dike C, Yehyawi N, Lysaker P, Dustin J, Caligiuri M, Lohr J, Lahiri DK, Nurnberger JI, Faraone SV, Geyer MA, Tsuang MT, Schork NJ, Salomon DR, Niculescu AB. Identification of blood biomarkers for psychosis using convergent functional genomics. Mol Psychiatry 2011; 16:37-58. [PMID: 19935739 DOI: 10.1038/mp.2009.117] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There are to date no objective clinical laboratory blood tests for psychotic disease states. We provide proof of principle for a convergent functional genomics (CFG) approach to help identify and prioritize blood biomarkers for two key psychotic symptoms, one sensory (hallucinations) and one cognitive (delusions). We used gene expression profiling in whole blood samples from patients with schizophrenia and related disorders, with phenotypic information collected at the time of blood draw, then cross-matched the data with other human and animal model lines of evidence. Topping our list of candidate blood biomarkers for hallucinations, we have four genes decreased in expression in high hallucinations states (Fn1, Rhobtb3, Aldh1l1, Mpp3), and three genes increased in high hallucinations states (Arhgef9, Phlda1, S100a6). All of these genes have prior evidence of differential expression in schizophrenia patients. At the top of our list of candidate blood biomarkers for delusions, we have 15 genes decreased in expression in high delusions states (such as Drd2, Apoe, Scamp1, Fn1, Idh1, Aldh1l1), and 16 genes increased in high delusions states (such as Nrg1, Egr1, Pvalb, Dctn1, Nmt1, Tob2). Twenty-five of these genes have prior evidence of differential expression in schizophrenia patients. Predictive scores, based on panels of top candidate biomarkers, show good sensitivity and negative predictive value for detecting high psychosis states in the original cohort as well as in three additional cohorts. These results have implications for the development of objective laboratory tests to measure illness severity and response to treatment in devastating disorders such as schizophrenia.
Collapse
Affiliation(s)
- S M Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Zhang W, Chen X, Gong W, Tang J, Tan L, Guo H, Yao YG. Common promoter variants of the NDUFV2 gene do not confer susceptibility to schizophrenia in Han Chinese. Behav Brain Funct 2010; 6:75. [PMID: 21190551 PMCID: PMC3022841 DOI: 10.1186/1744-9081-6-75] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 12/29/2010] [Indexed: 12/14/2022] Open
Abstract
Background The NADH-ubiquinone oxidoreductase flavoprotein gene (NDUFV2), which encodes a 24 kD mitochondrial complex I subunit, has been reported to be positively associated with schizophrenia and bipolar disorder in different populations. Methods We genotyped the promoter variants of this gene (rs6506640 and rs1156044) by direct sequencing in 529 unrelated Han Chinese schizophrenia patients and 505 matched controls. Fisher's Exact test was performed to assess whether these two reported single nucleotide polymorphisms (SNPs) confer susceptibility to schizophrenia in Chinese. Results Allele, genotype and haplotype comparison between the case and control groups showed no statistical significance, suggesting no association between the NDUFV2 gene promoter variants and schizophrenia in Han Chinese. Conclusion The role of NDUFV2 played in schizophrenia needs to be further studied. Different racial background and/or population substructure might account for the inconsistent results between studies.
Collapse
Affiliation(s)
- Wen Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | | | | | | | | | | | | |
Collapse
|
183
|
Talkowski ME, McCann KL, Chen M, McClain L, Bamne M, Wood J, Chowdari KV, Watson A, Prasad KM, Kirov G, Georgieva L, Toncheva D, Mansour H, Lewis DA, Owen M, O’Donovan M, Papasaikas P, Sullivan P, Ruderfer D, Yao JK, Leonard S, Thomas P, Miyajima F, Quinn J, Lopez AJ, Nimgaonkar VL. Fine-mapping reveals novel alternative splicing of the dopamine transporter. Am J Med Genet B Neuropsychiatr Genet 2010; 153B:1434-47. [PMID: 20957647 PMCID: PMC4575812 DOI: 10.1002/ajmg.b.31125] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 08/04/2010] [Indexed: 01/14/2023]
Abstract
The dopamine transporter gene (SLC6A3, DAT) has been implicated in the pathogenesis of numerous psychiatric and neurodevelopmental disorders, including schizophrenia (SZ). We previously detected association between SZ and intronic SLC6A3 variants that replicated in two independent Caucasian samples, but had no obvious function. In follow-up analyses, we sequenced the coding and intronic regions of SLC6A3 to identify complete linkage disequilibrium patterns of common variations. We genotyped 78 polymorphisms, narrowing the potentially causal region to two correlated clusters of associated SNPs localized predominantly to introns 3 and 4. Our computational analysis of these intronic regions predicted a novel cassette exon within intron 3, designated E3b, which is conserved among primates. We confirmed alternative splicing of E3b in post-mortem human substantia nigra (SN). As E3b introduces multiple in-frame stop codons, the SLC6A3 open reading frame is truncated and the spliced product may undergo nonsense mediated decay. Thus, factors that increase E3b splicing could reduce the amount of unspliced product available for translation. Observations consistent with this prediction were made using cellular assays and in post-mortem human SN. In mini-gene constructs, the extent of splicing is also influenced by at least two common haplotypes, so the alternative splicing was evaluated in relation to SZ risk. Meta-analyses across genome-wide association studies did not support the initial associations and further post-mortem studies did not suggest case-control differences in splicing. These studies do not provide a compelling link to schizophrenia. However, the impact of the alternative splicing on other neuropsychiatric disorders should be investigated. © 2010 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Michael E. Talkowski
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Kathleen L. McCann
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Michael Chen
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Lora McClain
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Mikhil Bamne
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joel Wood
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kodavali V. Chowdari
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Annie Watson
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Konasale M. Prasad
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - George Kirov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, UK
| | - Lyudmilla Georgieva
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, UK
| | | | - Hader Mansour
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David A. Lewis
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Michael Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, UK
| | - Michael O’Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, UK
| | - Panagiotis Papasaikas
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Patrick Sullivan
- Department of Genetics & Carolina Center for Genome Science, University of North Carolina, Chapel Hill, North Carolina
| | - Douglas Ruderfer
- Center for Human Genetic Research, Massachusetts General Hospital and Broad Institute, Boston, Massachusetts
| | - Jeffrey K Yao
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Sherry Leonard
- Department of Psychiatry, University of Colorado at Denver, Aurora, Colorado
| | - Pramod Thomas
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Fabio Miyajima
- Division of Human Anatomy and Cell Biology School of Biomedical Sciences, University of Liverpool, Liverpool, UK
| | - John Quinn
- Division of Human Anatomy and Cell Biology School of Biomedical Sciences, University of Liverpool, Liverpool, UK
| | - A. Javier Lopez
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Vishwajit L. Nimgaonkar
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| |
Collapse
|
184
|
Epigenetic GABAergic targets in schizophrenia and bipolar disorder. Neuropharmacology 2010; 60:1007-16. [PMID: 21074545 DOI: 10.1016/j.neuropharm.2010.10.021] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 10/18/2010] [Accepted: 10/21/2010] [Indexed: 12/15/2022]
Abstract
It is becoming increasingly clear that a dysfunction of the GABAergic/glutamatergic network in telencephalic brain structures may be the pathogenetic mechanism underlying psychotic symptoms in schizophrenia (SZ) and bipolar (BP) disorder patients. Data obtained in Costa's laboratory (1996-2009) suggest that this dysfunction may be mediated primarily by a downregulation in the expression of GABAergic genes (e.g., glutamic acid decarboxylase₆₇[GAD₆₇] and reelin) associated with DNA methyltransferase (DNMT)-dependent hypermethylation of their promoters. A pharmacological strategy to reduce the hypermethylation of GABAergic promoters is to administer drugs, such as the histone deacetylase (HDAC) inhibitor valproate (VPA), that induce DNA-demethylation when administered at doses that facilitate chromatin remodeling. The benefits elicited by combining VPA with antipsychotics in the treatment of BP disorder suggest that an investigation of the epigenetic interaction of these drugs is warranted. Our studies in mice suggest that when associated with VPA, clinically relevant doses of clozapine elicit a synergistic potentiation of VPA-induced GABAergic promoter demethylation. Olanzapine and quetiapine (two clozapine congeners) also facilitate chromatin remodeling but at doses higher than used clinically, whereas haloperidol and risperidone are inactive. Hence, the synergistic potentiation of VPA's action on chromatin remodeling by clozapine appears to be a unique property of the dibenzepines and is independent of their action on catecholamine or serotonin receptors. By activating DNA-demethylation, the association of clozapine or its derivatives with VPA or other more potent and selective HDAC inhibitors may be considered a promising treatment strategy for normalizing GABAergic promoter hypermethylation and the GABAergic gene expression downregulation detected in the postmortem brain of SZ and BP disorder patients. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.
Collapse
|
185
|
Ribbe K, Friedrichs H, Begemann M, Grube S, Papiol S, Kästner A, Gerchen MF, Ackermann V, Tarami A, Treitz A, Flögel M, Adler L, Aldenhoff JB, Becker-Emner M, Becker T, Czernik A, Dose M, Folkerts H, Freese R, Günther R, Herpertz S, Hesse D, Kruse G, Kunze H, Franz M, Löhrer F, Maier W, Mielke A, Müller-Isberner R, Oestereich C, Pajonk FG, Pollmächer T, Schneider U, Schwarz HJ, Kröner-Herwig B, Havemann-Reinecke U, Frahm J, Stühmer W, Falkai P, Brose N, Nave KA, Ehrenreich H. The cross-sectional GRAS sample: a comprehensive phenotypical data collection of schizophrenic patients. BMC Psychiatry 2010; 10:91. [PMID: 21067598 PMCID: PMC3002316 DOI: 10.1186/1471-244x-10-91] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 11/10/2010] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Schizophrenia is the collective term for an exclusively clinically diagnosed, heterogeneous group of mental disorders with still obscure biological roots. Based on the assumption that valuable information about relevant genetic and environmental disease mechanisms can be obtained by association studies on patient cohorts of ≥ 1000 patients, if performed on detailed clinical datasets and quantifiable biological readouts, we generated a new schizophrenia data base, the GRAS (Göttingen Research Association for Schizophrenia) data collection. GRAS is the necessary ground to study genetic causes of the schizophrenic phenotype in a 'phenotype-based genetic association study' (PGAS). This approach is different from and complementary to the genome-wide association studies (GWAS) on schizophrenia. METHODS For this purpose, 1085 patients were recruited between 2005 and 2010 by an invariable team of traveling investigators in a cross-sectional field study that comprised 23 German psychiatric hospitals. Additionally, chart records and discharge letters of all patients were collected. RESULTS The corresponding dataset extracted and presented in form of an overview here, comprises biographic information, disease history, medication including side effects, and results of comprehensive cross-sectional psychopathological, neuropsychological, and neurological examinations. With >3000 data points per schizophrenic subject, this data base of living patients, who are also accessible for follow-up studies, provides a wide-ranging and standardized phenotype characterization of as yet unprecedented detail. CONCLUSIONS The GRAS data base will serve as prerequisite for PGAS, a novel approach to better understanding 'the schizophrenias' through exploring the contribution of genetic variation to the schizophrenic phenotypes.
Collapse
Affiliation(s)
- Katja Ribbe
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Heidi Friedrichs
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Martin Begemann
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Sabrina Grube
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Sergi Papiol
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany,DFG Research Center for Molecular Physiology of the Brain (CMPB), Germany
| | - Anne Kästner
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Martin F Gerchen
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Verena Ackermann
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Asieh Tarami
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Annika Treitz
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Marlene Flögel
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Lothar Adler
- Department of Psychiatry and Psychotherapy, Ecumenical Hospital Hainich, Germany
| | - Josef B Aldenhoff
- Hospital of Psychiatry and Psychotherapy, Center for Integrative Psychiatry, Kiel, Germany
| | - Marianne Becker-Emner
- Karl-Jaspers-Hospital, Psychiatric Federation Oldenburger Land, Bad Zwischenahn, Germany
| | - Thomas Becker
- Department of Psychiatry II, Ulm University, District Hospital Günzburg, Germany
| | - Adelheid Czernik
- Department of Psychiatry and Psychotherapy, Hospital Fulda, Germany
| | - Matthias Dose
- Department of Psychiatry and Psychotherapy, Isar-Amper-Hospital, Taufkirchen (Vils), Germany
| | - Here Folkerts
- Department of Psychiatry and Psychotherapy, Reinhard-Nieter Hospital, Wilhelmshaven, Germany
| | - Roland Freese
- Vitos Hospital of Forensic Psychiatry Eltville, Eltville, Germany
| | - Rolf Günther
- Vitos Hospital of Psychiatry and Psychotherapy Merxhausen, Kassel, Germany
| | - Sabine Herpertz
- Department of Psychiatry and Psychotherapy, University of Rostock, Germany
| | - Dirk Hesse
- Hospital of Forensic Psychiatry, Moringen, Germany
| | - Gunther Kruse
- Hospital of Psychiatry and Psychotherapy Langenhagen, Regional Hospitals Hannover, Germany
| | - Heinrich Kunze
- Vitos Hospital of Psychiatry and Psychotherapy, Bad Emstal-Merxhausen, Germany
| | - Michael Franz
- Vitos Hospital of Psychiatry and Psychotherapy, Bad Emstal-Merxhausen, Germany
| | - Frank Löhrer
- Addiction Hospital "Am Waldsee", Rieden, Germany
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, University Medical Center of Bonn, Germany
| | - Andreas Mielke
- Vitos Hospital of Psychiatry and Psychotherapy Merxhausen, Hofgeismar, Germany
| | | | - Cornelia Oestereich
- Department of Psychiatry and Psychotherapy, Regional Hospitals Hannover, Wunstorf, Germany
| | | | - Thomas Pollmächer
- Department of Psychiatry and Psychotherapy, Hospital Ingolstadt, Germany
| | - Udo Schneider
- Department of Psychiatry and Psychotherapy, Hospital Lübbecke, Germany
| | | | | | - Ursula Havemann-Reinecke
- Department of Psychiatry and Psychotherapy, University Medical Center of Göttingen, Germany,DFG Research Center for Molecular Physiology of the Brain (CMPB), Germany
| | - Jens Frahm
- Biomedical NMR Research GmbH, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany,DFG Research Center for Molecular Physiology of the Brain (CMPB), Germany,Founders of the GRAS Initiative
| | - Walter Stühmer
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute of Experimental Medicine, Göttingen, Germany,DFG Research Center for Molecular Physiology of the Brain (CMPB), Germany,Founders of the GRAS Initiative
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Medical Center of Göttingen, Germany,DFG Research Center for Molecular Physiology of the Brain (CMPB), Germany,Founders of the GRAS Initiative
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany,DFG Research Center for Molecular Physiology of the Brain (CMPB), Germany,Founders of the GRAS Initiative
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany,DFG Research Center for Molecular Physiology of the Brain (CMPB), Germany,Founders of the GRAS Initiative
| | - Hannelore Ehrenreich
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany,DFG Research Center for Molecular Physiology of the Brain (CMPB), Germany,Founders of the GRAS Initiative
| |
Collapse
|
186
|
Tiwari AK, Zai CC, Müller DJ, Kennedy JL. Genetics in schizophrenia: where are we and what next? DIALOGUES IN CLINICAL NEUROSCIENCE 2010. [PMID: 20954426 PMCID: PMC3181975 DOI: 10.31887/dcns.2010.12.3/atiwari] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Understanding the genetic basis of schizophrenia continues to be major challenge. The research done during the last two decades has provided several candidate genes which unfortunately have not been consistently replicated across or within a population. The recent genome-wide association studies (GWAS) and copy number variation (CNV) studies have provided important evidence suggesting a role of both common and rare large CNVs in schizophrenia genesis. The burden of rare copy number variations appears to be increased in schizophrenia patients. A consistent observation among the GWAS studies is the association with schizophrenia of genetic markers in the major histocompatibility complex (6p22.1)-containing genes including NOTCH4 and histone protein loci. Molecular genetic studies are also demonstrating that there is more overlap between the susceptibility genes for schizophrenia and bipolar disorder than previously suspected. In this review we summarize the major findings of the past decade and suggest areas of future research.
Collapse
Affiliation(s)
- Arun K Tiwari
- Neurogenetics section, Neuroscience department, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
187
|
Greenbaum L, Alkelai A, Rigbi A, Kohn Y, Lerer B. Evidence for association of the GLI2
gene with tardive dyskinesia in patients with chronic schizophrenia. Mov Disord 2010; 25:2809-17. [DOI: 10.1002/mds.23377] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
188
|
Genetic repositories for the study of major psychiatric conditions: what do we know about ethnic minorities' genetic vulnerability? Mol Psychiatry 2010; 15:970-5. [PMID: 20177407 DOI: 10.1038/mp.2010.11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In spite of considerable efforts, no genes of major effect have been found across an entire diagnostic category in psychiatry. Possible reasons for this may include difficulties in defining the phenotype, the complex relationship between genotype and gene expression and population stratification. This last problem has often been managed by restricting genetic sampling to only one ethnic group. An unintended consequence of using this strategy is that the major repositories of genetic material for the study of psychiatric conditions in the United States suffer from a paucity of genetic samples from non-Caucasian groups. Thus, these groups are being relatively understudied in terms of the genetic antecedents to psychiatric disease. The authors provide solutions including the need to augment the representation of African-American, Latino and Asian-Americans among research participants; a more nuanced approach to identify ancestry; and the development of analytic and genetic strategies to handle the issue of ethnic heterogeneity in samples.
Collapse
|
189
|
Huang J, Perlis RH, Lee PH, Rush AJ, Fava M, Sachs GS, Lieberman J, Hamilton SP, Sullivan P, Sklar P, Purcell S, Smoller JW. Cross-disorder genomewide analysis of schizophrenia, bipolar disorder, and depression. Am J Psychiatry 2010; 167:1254-63. [PMID: 20713499 PMCID: PMC3880556 DOI: 10.1176/appi.ajp.2010.09091335] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Family and twin studies indicate substantial overlap of genetic influences on psychotic and mood disorders. Linkage and candidate gene studies have also suggested overlap across schizophrenia, bipolar disorder, and major depressive disorder. The purpose of this study was to apply genomewide association study (GWAS) analysis to address the specificity of genetic effects on these disorders. METHOD The authors combined GWAS data from three large effectiveness studies of schizophrenia (CATIE, genotyped: N=741), bipolar disorder (STEP-BD, geno-typed: N=1,575), and major depressive disorder (STAR*D, genotyped: N=1,938) as well as from psychiatrically screened control subjects (NIMH-Genetics Repository: N=1,204). A two-stage analytic procedure involving an omnibus test of allele frequency differences among case and control groups was applied, followed by a model selection step to identify the best-fitting model of allelic effects across disorders. RESULTS The strongest result was seen for a single nucleotide polymorphism near the adrenomedullin (ADM) gene (rs6484218), with the best-fitting model indicating that the effect was specific to bipolar II disorder. Findings also revealed evidence suggesting that several genes may have effects that transcend clinical diagnostic boundaries, including variants in NPAS3 that showed pleiotropic effects across schizophrenia, bipolar disorder, and major depressive disorder. CONCLUSIONS This study provides the first genomewide significant evidence implicating variants near the ADM gene on chromosome 11p15 in psychopathology, with effects that appear to be specific to bipolar II disorder. Although genomewide significant evidence of cross-disorder effects was not detected, the results provide evidence that there are both pleiotropic and disorder-specific effects on major mental illness and illustrate an approach to dissecting the genetic basis of mood and psychotic disorders that can inform future large-scale cross-disorder GWAS analyses.
Collapse
Affiliation(s)
- Jie Huang
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, and Psychiatric Genetics Program in Mood and Anxiety Disorders, Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Psychiatric Genetics Program in Mood and Anxiety Disorders, Massachusetts General Hospital, Boston, MA
| | - Roy H. Perlis
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, and Psychiatric Genetics Program in Mood and Anxiety Disorders, Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA
| | - Phil H. Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, and Psychiatric Genetics Program in Mood and Anxiety Disorders, Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Psychiatric Genetics Program in Mood and Anxiety Disorders, Massachusetts General Hospital, Boston, MA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA
| | | | - Maurizio Fava
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Gary S. Sachs
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Jeffrey Lieberman
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University and New York State Psychiatric Institute, New York, NY
| | - Steven P. Hamilton
- Department of Psychiatry and Institute for Human Genetics, University of California, San Francisco, CA
| | - Patrick Sullivan
- Departments of Genetics, Psychiatry, and Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pamela Sklar
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, and Psychiatric Genetics Program in Mood and Anxiety Disorders, Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA
| | - Shaun Purcell
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, and Psychiatric Genetics Program in Mood and Anxiety Disorders, Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA
| | - Jordan W. Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, and Psychiatric Genetics Program in Mood and Anxiety Disorders, Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Psychiatric Genetics Program in Mood and Anxiety Disorders, Massachusetts General Hospital, Boston, MA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA
| |
Collapse
|
190
|
Bloss CS, Schiabor KM, Schork NJ. Human behavioral informatics in genetic studies of neuropsychiatric disease: multivariate profile-based analysis. Brain Res Bull 2010; 83:177-88. [PMID: 20433907 PMCID: PMC2941546 DOI: 10.1016/j.brainresbull.2010.04.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 04/17/2010] [Accepted: 04/21/2010] [Indexed: 01/23/2023]
Abstract
While genome-wide association (GWA) studies have yielded notable findings with regard to the identification of risk variants in diseases such as obesity and diabetes, similar studies of schizophrenia - and neuropsychiatric diseases in general - have failed to produce strong findings. One, plausible explanation for this relates to phenotypic heterogeneity and what may be inherent imprecision associated with diagnostic categories in neuropsychiatric disorders. In this review we discuss a general approach to addressing the problem of heterogeneity that draws on concepts in behavioral informatics and the use of multivariable behavioral profiles in genetic studies of neuropsychiatric disease. The use of behavioral profiles as phenotypes eliminates the need for categorizing individuals with different 'subtypes' of a disease into one group and provides a way to investigate genetic susceptibility to different neuropsychiatric disorders that share similar clinical characteristics, such as schizophrenia and bipolar disorder. Further, behavioral profiles are a direct, quantitative representation of the emotional, personality, and neurocognitive functioning of the individuals being studied, and as such, the use of these profiles may provide increased statistical power to detect genetic associations and linkages. We describe and discuss four general data analysis approaches that can be used to analyze and integrate multivariate behavioral profile data and high-dimensional genomic data. Ultimately, we propose that behavioral profile-based phenotypes provide a meaningful alternative to the use of single measures, such as diagnostic category, in genetic association studies of neuropsychiatric disease.
Collapse
Affiliation(s)
- Cinnamon S. Bloss
- Scripps Genomic Medicine, Scripps Translational Science Institute, Scripps Health
| | - Kelly M. Schiabor
- Scripps Genomic Medicine, Scripps Translational Science Institute, Scripps Health
| | - Nicholas J. Schork
- Scripps Genomic Medicine, Scripps Translational Science Institute, Scripps Health
- Department of Molecular and Experimental Medicine, The Scripps Research Institute
| |
Collapse
|
191
|
Cytochrome P450 testing for prescribing antipsychotics in adults with schizophrenia: systematic review and meta-analyses. THE PHARMACOGENOMICS JOURNAL 2010; 11:1-14. [PMID: 20877299 DOI: 10.1038/tpj.2010.73] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
There is wide variability in the response of individuals to standard doses of antipsychotic drugs. It has been suggested that this may be partly explained by differences in the cytochrome P450 (CYP450) enzyme system responsible for metabolizing the drugs. We conducted a systematic review and meta-analyses to consider whether testing for CYP450 single nucleotide polymorphisms in adults starting antipsychotic treatment for schizophrenia predicts and leads to improvements in clinical outcomes. High analytic validity in terms of sensitivity and specificity was seen in studies reporting P450 testing. However, there was limited evidence of the role of CYP2D6 polymorphisms in antipsychotic efficacy, although there was an association between CYP2D6 genotype and extrapyramidal adverse effects. No studies reported on the prospective use of CYP2D6 genotyping tests in clinical practice. In conclusion, evidence of clinical validity and utility of CYP2D6 testing in patients being prescribed antipsychotics is lacking, and thus, routine pharmacogenetic testing prior to antipsychotic prescription cannot be supported at present. Further research is required to improve the evidence base and to generate data on clinical validity and clinical utility.
Collapse
|
192
|
Benke KS, Fallin MD. Methods: genetic epidemiology. Clin Lab Med 2010; 30:795-814. [PMID: 20832653 DOI: 10.1016/j.cll.2010.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Given the potential benefits of gene identification in psychiatry, genetic epidemiology has become a mainstream discipline within the field. This article discusses the main tools for gene discovery. The focus is on the designs and analytic approaches for each of these methods. Because most gene discovery has now moved to genetic association studies, and most recently to genome-wide association studies, the focus is on methods for this design. Also highlighted are the current challenges of genetic epidemiology as a prelude to future approaches that may be applied to psychiatric disorders in the coming years.
Collapse
Affiliation(s)
- Kelly S Benke
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, W6033, Baltimore, MD 21205, USA
| | | |
Collapse
|
193
|
Stergiakouli E, Thapar A. Fitting the pieces together: current research on the genetic basis of attention-deficit/hyperactivity disorder (ADHD). Neuropsychiatr Dis Treat 2010; 6:551-60. [PMID: 20856918 PMCID: PMC2938304 DOI: 10.2147/ndt.s11322] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly disruptive childhood-onset disorder that often persists into adolescence and adulthood. Comorbidity with other problems, such as autism, dyslexia and conduct disorder (CD) is very common. Although little is known about the pathophysiology of ADHD, family, twin and adoption studies have shown that it is highly heritable. Whole genome linkage studies suggest there are no common susceptibility genes of moderate effect size. Most published research has been based on functional candidate gene studies. The most consistent evidence for association with ADHD relates to a dopamine D4 receptor (DRD4) gene variable number tandem repeat (VNTR), a dopamine D5 receptor (DRD5) gene microsatellite and a dopamine transporter (DAT1) gene VNTR. In addition, the catechol-O-methyltransferase (COMT) val158/108 met variant has been shown to increase risk for associated antisocial behavior. The first genome-wide association studies (GWAS) of ADHD have been completed and although larger studies are still required to detect common risk variants, novel risk pathways are being suggested for ADHD. Further research on the contribution of rare variants, larger genome-wide association and sequencing studies and ADHD phenotype refinement is now needed.
Collapse
Affiliation(s)
- Evangelia Stergiakouli
- Department of Psychological, Medicine and Neurology, MRC Centre, for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff, University, United Kingdom
| | | |
Collapse
|
194
|
Donohoe G, Morris DW, Corvin A. The psychosis susceptibility gene ZNF804A: associations, functions, and phenotypes. Schizophr Bull 2010; 36:904-9. [PMID: 20688871 PMCID: PMC2930340 DOI: 10.1093/schbul/sbq080] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
As the first gene to have achieved genome-wide significance for psychosis, ZNF804A has predictably been a subject of intense research activity. We review the evidence to date for the association between schizophrenia and the original risk variant rs1344706 identified as well as additional common and rare variants at this locus. We describe the still scant literature on the biological function of ZNF804A and discuss the efforts being made to characterize and refine the associated phenotype using imaging and neuropsychological approaches. We conclude that ZNF804A is robustly, if modestly, associated with schizophrenia risk, with much work still remaining to elucidate its role in schizophrenia biology.
Collapse
Affiliation(s)
- Gary Donohoe
- Department of Psychiatry, Trinity College Dublin, Trinity Health Sciences Building, St. James's Hospital, Dublin 8, Ireland.
| | - Derek W. Morris
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity College Dublin, Trinity Health Sciences Building, St. James’s Hospital, Dublin 8, Ireland,Institute of Molecular Medicine, Trinity College Dublin, Ireland
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity College Dublin, Trinity Health Sciences Building, St. James’s Hospital, Dublin 8, Ireland,Institute of Molecular Medicine, Trinity College Dublin, Ireland,Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland
| |
Collapse
|
195
|
Jia P, Wang L, Meltzer HY, Zhao Z. Common variants conferring risk of schizophrenia: a pathway analysis of GWAS data. Schizophr Res 2010; 122:38-42. [PMID: 20659789 PMCID: PMC2933424 DOI: 10.1016/j.schres.2010.07.001] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Revised: 06/26/2010] [Accepted: 07/01/2010] [Indexed: 01/01/2023]
Abstract
Unlike the typical analysis of single markers in genome-wide association studies (GWAS), we incorporated Gene Set Enrichment Analysis (GSEA) and hypergeometric test and combined them using Fisher's combined method to perform pathway-based analysis in order to detect genes' combined effects on mediating schizophrenia. A few pathways were consistently found to be top ranked and likely associated with schizophrenia by these methods; they are related to metabolism of glutamate, the process of apoptosis, inflammation, and immune system (e.g., glutamate metabolism pathway, TGF-beta signaling pathway, and TNFR1 pathway). The genes involved in these pathways had not been detected by single marker analysis, suggesting this approach may complement the original analysis of GWAS dataset.
Collapse
Affiliation(s)
- Peilin Jia
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Psychiatry, Vanderbilt University, Nashville, TN 37232, USA
| | - Lily Wang
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Herbert Y Meltzer
- Department of Psychiatry, Vanderbilt University, Nashville, TN 37232, USA
| | - Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Psychiatry, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
196
|
Athanasiu L, Mattingsdal M, Kähler AK, Brown A, Gustafsson O, Agartz I, Giegling I, Muglia P, Cichon S, Rietschel M, Pietiläinen OP, Peltonen L, Bramon E, Collier D, St. Clair D, Sigurdsson E, Petursson H, Rujescu D, Melle I, Steen VM, Djurovic S, Andreassen OA. Gene variants associated with schizophrenia in a Norwegian genome-wide study are replicated in a large European cohort. J Psychiatr Res 2010; 44:748-53. [PMID: 20185149 PMCID: PMC3224994 DOI: 10.1016/j.jpsychires.2010.02.002] [Citation(s) in RCA: 174] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 02/01/2010] [Accepted: 02/02/2010] [Indexed: 02/03/2023]
Abstract
We have performed a genome-wide association study (GWAS) of schizophrenia in a Norwegian discovery sample of 201 cases and 305 controls (TOP study) with a focused replication analysis in a larger European sample of 2663 cases and 13,780 control subjects (SGENE-plus study). Firstly, the discovery sample was genotyped with Affymetrix Genome-Wide Human SNP Array 6.0 and 572,888 markers were tested for schizophrenia association. No SNPs in the discovery sample attained genome-wide significance (P<8.7 x 10(-8)). Secondly, based on the GWAS data, we selected 1000 markers with the lowest P values in the discovery TOP sample, and tested these (or HapMap-based surrogates) for association in the replication sample. Sixteen loci were associated with schizophrenia (nominal P value<0.05 and concurring OR) in the replication sample. As a next step, we performed a combined analysis of the findings from these two studies, and the strongest evidence for association with schizophrenia was provided for markers rs7045881 on 9p21, rs433598 on 16p12 and rs10761482 on 10q21. The markers are located in PLAA, ACSM1 and ANK3, respectively. PLAA has not previously been described as a susceptibility gene, but 9p21 is implied as a schizophrenia linkage region. ACSM1 has been identified as a susceptibility gene in a previous schizophrenia GWAS study. The association of ANK3 with schizophrenia is intriguing in light of recent associations of ANK3 with bipolar disorder, thereby supporting the hypothesis of an overlap in genetic susceptibility between these psychopathological entities.
Collapse
Affiliation(s)
- Lavinia Athanasiu
- Institute of Psychiatry, University of Oslo, P.O. 1130, Blindern, N-0318 Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Ulleval, Kirkeveien 166, N-0407 Oslo, Norway
- Department of Psychiatry, Oslo University Hospital, Ulleval, Kirkeveien 166, N-0407 Oslo, Norway
| | - Morten Mattingsdal
- Institute of Psychiatry, University of Oslo, P.O. 1130, Blindern, N-0318 Oslo, Norway
- Bioinformatics Core Facility, Institute of Medical Informatics, Oslo University Hospital, Montebello 0310, Norway
| | - Anna K. Kähler
- Institute of Psychiatry, University of Oslo, P.O. 1130, Blindern, N-0318 Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Ulleval, Kirkeveien 166, N-0407 Oslo, Norway
- Department of Psychiatry, Oslo University Hospital, Ulleval, Kirkeveien 166, N-0407 Oslo, Norway
| | - Andrew Brown
- Institute of Psychiatry, University of Oslo, P.O. 1130, Blindern, N-0318 Oslo, Norway
- Department of Biostatistics, University of Oslo, Blindern, N-0318 Oslo, Norway
- Department of Mathematics, University of Oslo, Blindern, N-0318 Oslo, Norway
| | - Omar Gustafsson
- Department of Psychiatry, Oslo University Hospital, Ulleval, Kirkeveien 166, N-0407 Oslo, Norway
| | - Ingrid Agartz
- Institute of Psychiatry, University of Oslo, P.O. 1130, Blindern, N-0318 Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Postboks 85, Vinderen, N-0319 Oslo, Norway
| | - Ina Giegling
- Division of Molecular and Clinical Neurobiology, Ludwig-Maximilians-University, Munich, Germany
| | | | - Sven Cichon
- Institute of Human Genetics, Department of Genomics, Life and Brain Centre, University of Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Centre Juelich, D-52425 Juelich, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Olli P.H. Pietiläinen
- Department for Molecular Medicine, National Public Health Institute, Helsinki, Finland
| | - Leena Peltonen
- Department for Molecular Medicine, National Public Health Institute, Helsinki, Finland
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- The Broad Institute, Cambridge, MA, USA
| | - Elvira Bramon
- Division of Psychological Medicine, Institute of Psychiatry, King’s College, London, UK
| | - David Collier
- Division of Psychological Medicine, Institute of Psychiatry, King’s College, London, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King’s College, London, UK
| | - David St. Clair
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Engilbert Sigurdsson
- Department of General Adult Psychiatry, Landspitali University Hospital, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Hannes Petursson
- Department of Psychiatry, Ludwig-Maximilians-University, Munich, Germany
| | - Dan Rujescu
- Department of Psychiatry, Ludwig-Maximilians-University, Munich, Germany
| | - Ingrid Melle
- Institute of Psychiatry, University of Oslo, P.O. 1130, Blindern, N-0318 Oslo, Norway
- Department of Psychiatry, Oslo University Hospital, Ulleval, Kirkeveien 166, N-0407 Oslo, Norway
| | - Vidar M. Steen
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Clinical Medicine, University of Bergen, Norway
- Centre for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Srdjan Djurovic
- Institute of Psychiatry, University of Oslo, P.O. 1130, Blindern, N-0318 Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Ulleval, Kirkeveien 166, N-0407 Oslo, Norway
- Department of Psychiatry, Oslo University Hospital, Ulleval, Kirkeveien 166, N-0407 Oslo, Norway
| | - Ole A. Andreassen
- Institute of Psychiatry, University of Oslo, P.O. 1130, Blindern, N-0318 Oslo, Norway
- Department of Psychiatry, Oslo University Hospital, Ulleval, Kirkeveien 166, N-0407 Oslo, Norway
| |
Collapse
|
197
|
Rivero O, Reif A, Sanjuán J, Moltó MD, Kittel-Schneider S, Nájera C, Töpner T, Lesch KP. Impact of the AHI1 gene on the vulnerability to schizophrenia: a case-control association study. PLoS One 2010; 5:e12254. [PMID: 20805890 PMCID: PMC2923617 DOI: 10.1371/journal.pone.0012254] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 07/15/2010] [Indexed: 01/13/2023] Open
Abstract
Background The Abelson helper integration-1 (AHI1) gene is required for both cerebellar and cortical development in humans. While the accelerated evolution of AHI1 in the human lineage indicates a role in cognitive (dys)function, a linkage scan in large pedigrees identified AHI1 as a positional candidate for schizophrenia. To further investigate the contribution of AHI1 to the susceptibility of schizophrenia, we evaluated the effect of AHI1 variation on the vulnerability to psychosis in two samples from Spain and Germany. Methodology/Principal Findings 29 single-nucleotide polymorphisms (SNPs) located in a genomic region including the AHI1 gene were genotyped in two samples from Spain (280 patients with psychotic disorders; 348 controls) and Germany (247 patients with schizophrenic disorders; 360 controls). Allelic, genotypic and haplotype frequencies were compared between cases and controls in both samples separately, as well as in the combined sample. The effect of genotype on several psychopathological measures (BPRS, KGV, PANSS) assessed in a Spanish subsample was also evaluated. We found several significant associations in the Spanish sample. Particularly, rs7750586 and rs911507, both located upstream of the AHI1 coding region, were found to be associated with schizophrenia in the analysis of genotypic (p = 0.0033, and 0.031, respectively) and allelic frequencies (p = 0.001 in both cases). Moreover, several other risk and protective haplotypes were detected (0.006<p<0.036). Joint analysis also supported the association of rs7750586 and rs911507 with the risk for schizophrenia. The analysis of clinical measures also revealed an effect on symptom severity (minimum P value = 0.0037). Conclusions/Significance Our data support, in agreement with previous reports, an effect of AHI1 variation on the susceptibility to schizophrenia in central and southern European populations.
Collapse
Affiliation(s)
- Olga Rivero
- Unit of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
198
|
Lymphoblast and brain expression of AHI1 and the novel primate-specific gene, C6orf217, in schizophrenia and bipolar disorder. Schizophr Res 2010; 120:159-66. [PMID: 20452750 DOI: 10.1016/j.schres.2010.03.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 03/26/2010] [Accepted: 03/30/2010] [Indexed: 11/22/2022]
Abstract
Association with schizophrenia of the Abelson Helper Integration Site 1 (AHI1) gene on chromosome 6q23 and the adjacent primate-specific gene, C6orf217, was demonstrated in an inbred, Arab Israeli family sample and replicated in an Icelandic case control sample. Further support was provided by a second replication in a large European sample and a meta-analysis that supported association with schizophrenia of all seven alleles overtransmitted to affected subjects in the original study. We examined constitutive expression of AHI1 and C6orf217 in immortalized lymphoblasts of patients from the Arab Israeli family sample in which the association with schizophrenia was originally discovered and population-matched normal controls, and in post-mortem brain of patients with schizophrenia and bipolar (BP) disorder and control subjects from the Stanley Medical Research Institute Collection. We found a significant effect of diagnostic group in the lymphoblast sample (F=5.72; df=2,39; p=0.006). Patients with early age of onset had higher AHI1 expression than controls and later onset patients (p=0.002; 0.03 respectively). C6orf217 expression in lymphoblasts was too low to measure. We found no difference in brain expression of AHI1 in schizophrenia or BP patients compared to controls. However, there was a genotypic difference in AHI1 expression for SNP rs9321501, which was strongly associated with schizophrenia in the original study. Genotypes that included the undertransmitted C allele (CC/AC) showed lower expression than the homozygous AA genotype (F=4.73, df=2,83; p=0.028). There was no significant difference in brain expression of C6orf217 between patients and controls and no genotypic effect. This study provides further evidence for involvement of AHI1 in susceptibility to schizophrenia.
Collapse
|
199
|
Rodríguez-Murillo L, Lieberman JA. Una firma genética para las enfermedades psiquiátricas complejas. REVISTA DE PSIQUIATRIA Y SALUD MENTAL 2010; 3:75-8. [DOI: 10.1016/j.rpsm.2010.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
200
|
Konneker TI, Crowley JJ, Quackenbush CR, Keefe RSE, Perkins DO, Stroup TS, Lieberman JA, van den Oord E, Sullivan PF. No association of the serotonin transporter polymorphisms 5-HTTLPR and RS25531 with schizophrenia or neurocognition. Am J Med Genet B Neuropsychiatr Genet 2010; 153B:1115-7. [PMID: 20468059 PMCID: PMC3181051 DOI: 10.1002/ajmg.b.31077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A promoter polymorphism in the serotonin transporter gene has been widely studied in neuropsychiatry. We genotyped the 5-HTTLPR/rs25531 triallelic polymorphism in 728 schizophrenia cases from the CATIE study and 724 control subjects. In a logistic regression with case/control status as dependent variable and 7 ancestry-informative principal components as covariates, the effect of 5-HTTLPR/rs25531 composite genotype was not significant (odds ratio = 1.008, 95% CI 0.868-1.172, P = 0.91). In cases only, 5-HTTLPR/rs25531 was not associated with neurocognition (summary neurocognitive index P = 0.21, working memory P = 0.32) or symptomatology (PANSS positive P = 0.67 and negative symptoms P = 0.46). We were unable to identify association of the triallelic 5-HTTLPR with schizophrenia, neurocognition, or core psychotic symptoms even at levels of significance unadjusted for multiple comparisons.
Collapse
Affiliation(s)
- Thomas I Konneker
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7264, USA
| | | | | | | | | | | | | | | | | |
Collapse
|