51
|
Azimi T, Ghafouri-Fard S, Davood Omrani M, Mazdeh M, Arsang-Jang S, Sayad A, Taheri M. Vaccinia Related Kinase 2 (VRK2) expression in neurological disorders: schizophrenia, epilepsy and multiple sclerosis. Mult Scler Relat Disord 2017; 19:15-19. [PMID: 29100046 DOI: 10.1016/j.msard.2017.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 09/19/2017] [Accepted: 10/23/2017] [Indexed: 01/19/2023]
Abstract
BACKGROUND Schizophrenia (SCZ), epilepsy and Multiple Sclerosis (MS) are neurological disorders with increasing prevalence disturb the patients' lives and are regarded as burdens to the society. As multifactorial disorders, genetic susceptibility factors are involved in their pathogenesis. The Vaccinia-Related Kinase 2 (VRK2) gene codes for a serine threonine kinase recently reported to be contributed in the pathogenesis of some neurological disorders. In the present case-control study we compared the VRK2 gene expression in peripheral blood samples from SCZ, epilepsy and MS patients with normal subjects. METHOD A total of 300 subjects comprising 50 patients in each disease category (SCZ, epilepsy and MS) as well as 150 healthy individuals (50 matched controls for each disorder) participated in the current study. RESULT The VRK2 blood mRNA expression level was measured using the TaqMan real time PCR. The results demonstrated significant down-regulation of VRK2 gene in SCZ (P<0.0001), epilepsy (P=0.008) and MS (P=0.029) compared with the healthy subjects. CONCLUSION Consequently, VRK2 is suggested as a candidate gene for neurological disorders through its role in signaling pathway, the neuronal loss and stress response.
Collapse
Affiliation(s)
- Tahereh Azimi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, PO Box 1985717443, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, PO Box 1985717443, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, PO Box 1985717443, Tehran, Iran; Urogenital Stem Cell Research Center, Shahid Labbafi Nejad Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdokht Mazdeh
- Department of Neurology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahram Arsang-Jang
- Department of Epidemiology and Biostatistics, Faculty of Health, Qom University of Medical Sciences, Qom, Iran
| | - Arezou Sayad
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, PO Box 1985717443, Tehran, Iran.
| | - Mohammad Taheri
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, PO Box 1985717443, Tehran, Iran; Urogenital Stem Cell Research Center, Shahid Labbafi Nejad Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
52
|
van Os J, van der Steen Y, Islam MA, Gülöksüz S, Rutten BP, Simons CJ. Evidence that polygenic risk for psychotic disorder is expressed in the domain of neurodevelopment, emotion regulation and attribution of salience. Psychol Med 2017; 47:2421-2437. [PMID: 28436345 DOI: 10.1017/s0033291717000915] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The liability-threshold model of psychosis risk predicts stronger phenotypic manifestation of the polygenic risk score (PRS) in the healthy relatives of patients, as compared with healthy comparison subjects. METHODS First-degree relatives of patients with psychotic disorder (871 siblings and 812 parents) and healthy comparison subjects (n = 523) were interviewed three times in 6 years. Repeated measures of two psychosis phenotypes, the Community Assessment of Psychic Experiences (CAPE; self-report - subscales of positive, negative and depressive symptoms) and the Structured Interview for Schizotypy - Revised (SIS-R; clinical interview - subscales of positive and negative schizotypy), were examined for association with PRS. Interview-based lifetime rate of depressive and manic episodes were also examined, as was association with repeated measures of intelligence quotient (IQ). RESULTS In the relatives, PRS was associated with CAPE/SIS-R total score (respectively, B = 0.12, 95% CI 0.02-0.22 and B = 0.11, 95% CI 0.02-0.20), the SIS-R positive subscale (B = 0.16, 95% CI 0.04-0.28), the CAPE depression subscale (B = 0.21, 95% CI 0.07-0.34), any lifetime affective episode (OR 3.1, 95% CI 1.04-9.3), but not with IQ (B = -1.8, 95% CI -8.0 to 4.4). In the controls, similar associations were apparent between PRS on the one hand and SIS-R total score, SIS-R positive, SIS-R negative, any lifetime affective episode and, in contrast, lower IQ (B = -8.5, 95% CI -15.5 to -1.6). CONCLUSIONS In non-ill people, polygenic risk for psychotic disorder is expressed pleiotropically in the domain of neurodevelopment, emotion regulation and attribution of salience. In subjects at elevated genetic risk, emerging expression of neurodevelopmental alterations may create floor effects, obscuring genetic associations.
Collapse
Affiliation(s)
- J van Os
- Department of Psychiatry and Psychology,Maastricht University Medical Centre,Maastricht,The Netherlands
| | - Y van der Steen
- Department of Psychiatry and Psychology,Maastricht University Medical Centre,Maastricht,The Netherlands
| | - Md A Islam
- University of Groningen, University Medical Center Groningen, University Center for Psychiatry,Groningen,The Netherlands
| | - S Gülöksüz
- Department of Psychiatry and Psychology,Maastricht University Medical Centre,Maastricht,The Netherlands
| | - B P Rutten
- Department of Psychiatry and Psychology,Maastricht University Medical Centre,Maastricht,The Netherlands
| | - C J Simons
- Department of Psychiatry and Psychology,Maastricht University Medical Centre,Maastricht,The Netherlands
| |
Collapse
|
53
|
Ibrahim EC, Guillemot V, Comte M, Tenenhaus A, Zendjidjian XY, Cancel A, Belzeaux R, Sauvanaud F, Blin O, Frouin V, Fakra E. Modeling a linkage between blood transcriptional expression and activity in brain regions to infer the phenotype of schizophrenia patients. NPJ SCHIZOPHRENIA 2017; 3:25. [PMID: 28883405 PMCID: PMC5589880 DOI: 10.1038/s41537-017-0027-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/05/2017] [Accepted: 07/21/2017] [Indexed: 11/20/2022]
Abstract
Hundreds of genetic loci participate to schizophrenia liability. It is also known that impaired cerebral connectivity is directly related to the cognitive and affective disturbances in schizophrenia. How genetic susceptibility and brain neural networks interact to specify a pathological phenotype in schizophrenia remains elusive. Imaging genetics, highlighting brain variations, has proven effective to establish links between vulnerability loci and associated clinical traits. As previous imaging genetics works in schizophrenia have essentially focused on structural DNA variants, these findings could be blurred by epigenetic mechanisms taking place during gene expression. We explored the meaningful links between genetic data from peripheral blood tissues on one hand, and regional brain reactivity to emotion task assayed by blood oxygen level-dependent functional magnetic resonance imaging on the other hand, in schizophrenia patients and matched healthy volunteers. We applied Sparse Generalized Canonical Correlation Analysis to identify joint signals between two blocks of variables: (i) the transcriptional expression of 33 candidate genes, and (ii) the blood oxygen level-dependent activity in 16 region of interest. Results suggested that peripheral transcriptional expression is related to brain imaging variations through a sequential pathway, ending with the schizophrenia phenotype. Generalization of such an approach to larger data sets should thus help in outlining the pathways involved in psychiatric illnesses such as schizophrenia. IMAGING SEARCHING FOR LINKS TO AID DIAGNOSIS: Researchers explore links between the expression of genes associated with schizophrenia in blood cells and variations in brain activity during emotion processing. El Chérif Ibrahim and Eric Fakra at Aix-Marseille Université, France, and colleagues have developed a method to relate the expression levels of 33 schizophrenia susceptibility genes in blood cells and functional magnetic resonance imaging (fMRI) data obtained as individuals carry out a task that triggers emotional responses. Although they found no significant differences in the expression of genes between the 26 patients with schizophrenia and 26 healthy controls they examined, variations in activity in the superior temporal gyrus were strongly linked to schizophrenia-associated gene expression and presence of disease. Similar analyses of larger data sets will shed further light on the relationship between peripheral molecular changes and disease-related behaviors and ultimately, aid the diagnosis of neuropsychiatric disease.
Collapse
Affiliation(s)
- El Chérif Ibrahim
- Aix-Marseille Univ, CNRS, CRN2M, Marseille, France.
- Fondation FondaMental, Fondation de Recherche et de Soins en Santé Mentale, Créteil, France.
- Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France.
| | - Vincent Guillemot
- INSERM, U 1127, Paris, France
- CNRS, 7225, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMRS_1127, Paris, France
- ICM, Département des maladies du système nerveux and Département de Génétique, Hôpital Pitié-Salpêtrière, Paris, France
| | - Magali Comte
- Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France
| | - Arthur Tenenhaus
- Laboratoire des Signaux et Systèmes (L2S, UMR CNRS 8506), CentraleSupélec-CNRS Université Paris-Sud, Gif-sur-Yvette, France
- Bioinformatics/Biostatistics Platform IHU-A-ICM, Brain and Spine Institute, Paris, France
| | - Xavier Yves Zendjidjian
- Pôle Psychiatrie centre, Hôpital de la Conception, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Aida Cancel
- Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France
- Service Hospitalo-Universitaire de Psychiatrie Secteur Saint-Etienne, Hôpital Nord, Saint-Etienne, France
| | - Raoul Belzeaux
- Aix-Marseille Univ, CNRS, CRN2M, Marseille, France
- Fondation FondaMental, Fondation de Recherche et de Soins en Santé Mentale, Créteil, France
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Florence Sauvanaud
- Service Hospitalo-Universitaire de Psychiatrie Secteur Saint-Etienne, Hôpital Nord, Saint-Etienne, France
| | - Olivier Blin
- Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France
- CIC-UPCET et Pharmacologie Clinique, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | | | - Eric Fakra
- Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France.
- Service Hospitalo-Universitaire de Psychiatrie Secteur Saint-Etienne, Hôpital Nord, Saint-Etienne, France.
| |
Collapse
|
54
|
Need AC, Goldstein DB. Neuropsychiatric genomics in precision medicine: diagnostics, gene discovery, and translation. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 27757059 PMCID: PMC5067142 DOI: 10.31887/dcns.2016.18.3/aneed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Only a few years after its development, next-generation sequencing is rapidly becoming an essential part of clinical care for patients with serious neurological conditions, especially in the diagnosis of early-onset and severe presentations. Beyond this diagnostic role, there has been an explosion in definitive gene discovery in a range of neuropsychiatric diseases. This is providing new pointers to underlying disease biology and is beginning to outline a new framework for genetic stratification of neuropsychiatric disease, with clear relevance to both individual treatment optimization and clinical trial design. Here, we outline these developments and chart the expected impact on the treatment of neurological, neurodevelopmental, and psychiatric disease.
Collapse
Affiliation(s)
- Anna C Need
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, W12 ONN, UK
| | - David B Goldstein
- Institute for Genomic Medicine, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
55
|
Yue W, Yu X, Zhang D. Progress in genome-wide association studies of schizophrenia in Han Chinese populations. NPJ SCHIZOPHRENIA 2017; 3:24. [PMID: 28798405 PMCID: PMC5552785 DOI: 10.1038/s41537-017-0029-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/29/2017] [Accepted: 05/03/2017] [Indexed: 01/01/2023]
Abstract
Since 2006, genome-wide association studies of schizophrenia have led to the identification of numerous novel risk loci for this disease. However, there remains a geographical imbalance in genome-wide association studies, which to date have primarily focused on Western populations. During the last 6 years, genome-wide association studies in Han Chinese populations have identified both the sharing of susceptible loci across ethnicities and genes unique to Han Chinese populations. Here, we review recent progress in genome-wide association studies of schizophrenia in Han Chinese populations. Researchers have identified and replicated the sharing of susceptible genes, such as within the major histocompatibility complex, microRNA 137 (MIR137), zinc finger protein 804A (ZNF804A), vaccinia related kinase 2 (VRK2), and arsenite methyltransferase (AS3MT), across both European and East Asian populations. Several copy number variations identified in European populations have also been validated in the Han Chinese, including duplications at 16p11.2, 15q11.2-13.1, 7q11.23, and VIPR2 and deletions at 22q11.2, 1q21.1-q21.2, and NRXN1. However, these studies have identified some potential confounding factors, such as genetic heterogeneity and the effects of natural selection on tetraspanin 18 (TSPAN18) or zinc finger protein 323 (ZNF323), which may explain the population differences in genome-wide association studies. In the future, genome-wide association studies in Han Chinese populations should include meta-analyzes or mega-analyses with enlarged sample sizes across populations, deep sequencing, precision medicine treatment, and functional exploration of the risk genes for schizophrenia.
Collapse
Affiliation(s)
- Weihua Yue
- Institute of Mental Health, the Sixth Hospital, Peking University, 100191, Beijing, China.
- Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), 100191, Beijing, China.
| | - Xin Yu
- Institute of Mental Health, the Sixth Hospital, Peking University, 100191, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), 100191, Beijing, China
| | - Dai Zhang
- Institute of Mental Health, the Sixth Hospital, Peking University, 100191, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), 100191, Beijing, China
- Peking-Tsinghua Joint Center for Life Sciences & PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China
| |
Collapse
|
56
|
Yu H, Yan H, Li J, Li Z, Zhang X, Ma Y, Mei L, Liu C, Cai L, Wang Q, Zhang F, Iwata N, Ikeda M, Wang L, Lu T, Li M, Xu H, Wu X, Liu B, Yang J, Li K, Lv L, Ma X, Wang C, Li L, Yang F, Jiang T, Shi Y, Li T, Zhang D, Yue W. Common variants on 2p16.1, 6p22.1 and 10q24.32 are associated with schizophrenia in Han Chinese population. Mol Psychiatry 2017; 22:954-960. [PMID: 27922604 DOI: 10.1038/mp.2016.212] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 09/15/2016] [Accepted: 10/14/2016] [Indexed: 02/05/2023]
Abstract
Many schizophrenia susceptibility loci have been identified through genome-wide association studies (GWASs) in European populations. However, until recently, schizophrenia GWASs in non-European populations were limited to small sample sizes and have yielded few loci associated with schizophrenia. To identify genetic risk variations for schizophrenia in the Han Chinese population, we performed a two-stage GWAS of schizophrenia comprising 4384 cases and 5770 controls, followed by independent replications of 13 single-nucleotide polymorphisms in an additional 4339 schizophrenia cases and 7043 controls of Han Chinese ancestry. Furthermore, we conducted additional analyses based on the results in the discovery stage. The combined analysis confirmed evidence of genome-wide significant associations in the Han Chinese population for three loci, at 2p16.1 (rs1051061, in an exon of VRK2, P=1.14 × 10-12, odds ratio (OR)=1.17), 6p22.1 (rs115070292 in an intron of GABBR1, P=4.96 × 10-10, OR=0.77) and 10q24.32 (rs10883795 in an intron of AS3MT, P=7.94 × 10-10, OR=0.87; rs10883765 at an intron of ARL3, P=3.06 × 10-9, OR=0.87). The polygenic risk score based on Psychiatric Genomics Consortium schizophrenia GWAS data modestly predicted case-control status in the Chinese population (Nagelkerke R2: 1.7% ~5.7%). Our pathway analysis suggested that neurological biological pathways such as GABAergic signaling, dopaminergic signaling, cell adhesion molecules and myelination pathways are involved in schizophrenia. These findings provide new insights into the pathogenesis of schizophrenia in the Han Chinese population. Further studies are needed to establish the biological context and potential clinical utility of these findings.
Collapse
Affiliation(s)
- H Yu
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Department of Biochemistry, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health &National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
| | - H Yan
- Department of Biochemistry, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health &National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
| | - J Li
- Department of Biochemistry, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health &National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
| | - Z Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - X Zhang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Y Ma
- Department of Biochemistry, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health &National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
| | - L Mei
- Department of Biochemistry, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health &National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
| | - C Liu
- Department of Psychiatry, the University of Melbourne, Parkville, VIC, Australia
| | - L Cai
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Q Wang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, Psychiatric laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - F Zhang
- Department of Clinical Psychology, Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| | - N Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Aichi, Japan
| | - M Ikeda
- Department of Psychiatry, Fujita Health University School of Medicine, Aichi, Japan
| | - L Wang
- Department of Biochemistry, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health &National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
| | - T Lu
- Department of Biochemistry, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health &National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
| | - M Li
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, the Second Military Medical University, Shanghai, China
| | - H Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, the Second Military Medical University, Shanghai, China
| | - X Wu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, the Second Military Medical University, Shanghai, China
| | - B Liu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - J Yang
- Tianjin Anding Hospital, Tianjin, China
| | - K Li
- Hebei Mental Health Center, Baoding, Hebei, China
| | - L Lv
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - X Ma
- Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - C Wang
- Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - L Li
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - F Yang
- Beijing HuiLongGuan Hospital, Peking University, Beijing, China
| | - T Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Y Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - T Li
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, Psychiatric laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - D Zhang
- Department of Biochemistry, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health &National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
- Peking-Tsinghua Joint Center for Life Sciences/PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - W Yue
- Department of Biochemistry, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health &National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
| |
Collapse
|
57
|
Abstract
Epileptic encephalopathies represent a particularly severe form of epilepsy, associated with cognitive and behavioral deficits, including impaired social-communication and restricted, repetitive behaviors that are the hallmarks of autism spectrum disorder (ASD). With the advent of next-generation sequencing, the genetic landscape of epileptic encephalopathies is growing and demonstrates overlap with genes separately implicated in ASD. However, many questions remain about this connection, including whether epileptiform activity itself contributes to the development of ASD symptomatology. In this review, we compiled a database of genes associated with both epileptic encephalopathy and ASD, limiting our purview to Mendelian disorders not including inborn errors of metabolism, and we focused on the connection between ASD and epileptic encephalopathy rather than epilepsy broadly. Our review has four goals: to (1) discuss the overlapping presentations of ASD and monogenic epileptic encephalopathies; (2) examine the impact of the epilepsy itself on neurocognitive features, including ASD, in monogenic epileptic encephalopathies; (3) outline many of the genetic causes responsible for both ASD and epileptic encephalopathy; (4) provide an illustrative example of a final common pathway that may be implicated in both ASD and epileptic encephalopathy. We demonstrate that autistic features are a common association with monogenic epileptic encephalopathies. Certain epileptic encephalopathy syndromes, like infantile spasms, are especially linked to the development of ASD. The connection between seizures themselves and neurobehavioral deficits in these monogenic encephalopathies remains open to debate. Finally, advances in genetics have revealed many genes that overlap in ties to both ASD and epileptic encephalopathy and that play a role in diverse central nervous system processes. Increased attention to the autistic features of monogenic epileptic encephalopathies is warranted for both researchers and clinicians alike.
Collapse
Affiliation(s)
- Siddharth Srivastava
- Department of Neurology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA
| |
Collapse
|
58
|
Hill MJ, Killick R, Navarrete K, Maruszak A, McLaughlin GM, Williams BP, Bray NJ. Knockdown of the schizophrenia susceptibility gene TCF4 alters gene expression and proliferation of progenitor cells from the developing human neocortex. J Psychiatry Neurosci 2017; 42:181-188. [PMID: 27689884 PMCID: PMC5403663 DOI: 10.1503/jpn.160073] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Common variants in the TCF4 gene are among the most robustly supported genetic risk factors for schizophrenia. Rare TCF4 deletions and loss-of-function point mutations cause Pitt-Hopkins syndrome, a developmental disorder associated with severe intellectual disability. METHODS To explore molecular and cellular mechanisms by which TCF4 perturbation could interfere with human cortical development, we experimentally reduced the endogenous expression of TCF4 in a neural progenitor cell line derived from the developing human cerebral cortex using RNA interference. Effects on genome-wide gene expression were assessed by microarray, followed by Gene Ontology and pathway analysis of differentially expressed genes. We tested for genetic association between the set of differentially expressed genes and schizophrenia using genome-wide association study data from the Psychiatric Genomics Consortium and competitive gene set analysis (MAGMA). Effects on cell proliferation were assessed using high content imaging. RESULTS Genes that were differentially expressed following TCF4 knockdown were highly enriched for involvement in the cell cycle. There was a nonsignificant trend for genetic association between the differentially expressed gene set and schizophrenia. Consistent with the gene expression data, TCF4 knockdown was associated with reduced proliferation of cortical progenitor cells in vitro. LIMITATIONS A detailed mechanistic explanation of how TCF4 knockdown alters human neural progenitor cell proliferation is not provided by this study. CONCLUSION Our data indicate effects of TCF4 perturbation on human cortical progenitor cell proliferation, a process that could contribute to cognitive deficits in individuals with Pitt-Hopkins syndrome and risk for schizophrenia.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nicholas J. Bray
- Correspondence to: N. Bray, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University School of Medicine, Cardiff, UK;
| |
Collapse
|
59
|
Abstract
Schizophrenia (SZ) is a debilitating brain disorder with a complex genetic architecture. Genetic studies, especially recent genome-wide association studies (GWAS), have identified multiple variants (loci) conferring risk to SZ. However, how to efficiently extract meaningful biological information from bulk genetic findings of SZ remains a major challenge. There is a pressing need to integrate multiple layers of data from various sources, eg, genetic findings from GWAS, copy number variations (CNVs), association and linkage studies, gene expression, protein-protein interaction (PPI), co-expression, expression quantitative trait loci (eQTL), and Encyclopedia of DNA Elements (ENCODE) data, to provide a comprehensive resource to facilitate the translation of genetic findings into SZ molecular diagnosis and mechanism study. Here we developed the SZDB database (http://www.szdb.org/), a comprehensive resource for SZ research. SZ genetic data, gene expression data, network-based data, brain eQTL data, and SNP function annotation information were systematically extracted, curated and deposited in SZDB. In-depth analyses and systematic integration were performed to identify top prioritized SZ genes and enriched pathways. Multiple types of data from various layers of SZ research were systematically integrated and deposited in SZDB. In-depth data analyses and integration identified top prioritized SZ genes and enriched pathways. We further showed that genes implicated in SZ are highly co-expressed in human brain and proteins encoded by the prioritized SZ risk genes are significantly interacted. The user-friendly SZDB provides high-confidence candidate variants and genes for further functional characterization. More important, SZDB provides convenient online tools for data search and browse, data integration, and customized data analyses.
Collapse
Affiliation(s)
- Yong Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China;,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China;,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China;,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China,YGY and XJL are co-corresponding authors who jointly directed this work
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China;,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China;,YGY and XJL are co-corresponding authors who jointly directed this work
| |
Collapse
|
60
|
Cardoso MABS, do Nascimento TJ, Bernardo GP, Bernardo LP, Barbosa MMFL, Neto PJN, de Sousa DF, Júnior AGT, de Lima MAP, Moreira MM, de Sousa Gregório D, do Nascimento Santos LC, Rolim Neto ML. Are There Schizophrenia Genetic Markers and Mutations? A Systematic Review and Meta-Analyses. Health (London) 2017. [DOI: 10.4236/health.2017.95058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
61
|
Abstract
Epidemiological studies and mouse models suggest that maternal immune activation, induced clinically through prenatal exposure to one of several infectious diseases, is a risk factor in the development of schizophrenia. This is supported by the strong genetic association established by genome wide association studies (GWAS) between the human leukocyte antigen (HLA) locus and schizophrenia. HLA proteins (also known in mice as the major histocompatibility complex; MHC) are mediators of the T-lymphocyte responses, and genetic variability is well-established as a risk factor for autoimmune diseases and susceptibility to infectious diseases. Taken together, the findings strongly suggest that schizophrenia risk in a subgroup of patients is caused by an infectious disease, and/or an autoimmune phenomenon. However, this view may be overly simplistic. First, MHC proteins have a non-immune effect on synaptogenesis by modulating synaptic pruning by microglia and other mechanisms, suggesting that genetic variability could be compromising this physiological process. Second, some GWAS signals in the HLA locus map near non-HLA genes, such as the histone gene cluster. On the other hand, recent GWAS data show association signals near B-lymphocyte enhancers, which lend support for an infectious disease etiology. Thus, although the genetic findings implicating the HLA locus are very robust, how genetic variability in this region leads to schizophrenia remains to be elucidated.
Collapse
Affiliation(s)
- Ryan Mokhtari
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York, USA
| | - Herbert M Lachman
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York, USA; Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York, USA; Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York, USA; Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York, USA
| |
Collapse
|
62
|
Li J, Chen Z, Wang F, Ouyang Y, Zhang N, Yang M, Yan M, Zhu X, He X, Yuan D, Jin T. Polymorphisms of the TCF4 gene are associated with the risk of schizophrenia in the Han Chinese. Am J Med Genet B Neuropsychiatr Genet 2016; 171:1006-1012. [PMID: 27103199 DOI: 10.1002/ajmg.b.32449] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/15/2016] [Indexed: 01/30/2023]
Abstract
Schizophrenia (SCZ) is a complex and severe mental disorder with highly heritability (80%). Several large genome-wide association studies have identified that the transcription factor 4 (TCF4) polymorphisms were strongly associated with SCZ. Therefore, the present study was to replicate the potential relationships between the TCF4 polymorphisms and SCZ. Furthermore, the study also investigated whether other variants were associated with SCZ in the Han Chinese. We conducted a case-control study including 499 patients and 500 healthy controls. Five SNPs were successfully genotyped and evaluated the association with SCZ by using χ2 test and genetic model analysis. We found that the genotype "AG" of rs9320010 and "GA" of rs7235757 decreased SCZ risk (OR = 0.70, 95%CI = 0.50-0.99, P = 0.041; OR = 0.69, 95%CI = 0.49-0.97, P = 0.034, respectively). In the genetic model analysis, we also observed that the allele "A" of rs9320010 and "G" of rs7235757 were inversely related with the risk of SCZ in the dominant model (OR = 0.72, 95%CI = 0.52-0.98, P = 0.039; OR = 0.69, 95%CI = 0.50-0.96, P = 0.025, respectively). Further interaction and stratification analysis suggested that rs1452787 was notably correlated with increased SCZ risk in males (OR = 2.77, 95%CI = 1.43-5.35, P = 0.002). Our study indicated that rs9320010, rs7235757, and rs1452787 were prominently associated with SCZ. Further studies are required to verify our findings and focus on determining the biological functions of the SNPs. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jingjie Li
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Zhengshuai Chen
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Fengjiao Wang
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yongri Ouyang
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Ning Zhang
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Min Yang
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Mengdan Yan
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Xikai Zhu
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Xue He
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China.,Key Laboratory for Basic Life Science Research of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Dongya Yuan
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China.,Key Laboratory for Basic Life Science Research of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Tianbo Jin
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China.,Key Laboratory for Basic Life Science Research of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| |
Collapse
|
63
|
Liu J, Li M, Su B. GWAS-identified schizophrenia risk SNPs at TSPAN18 are highly diverged between Europeans and East Asians. Am J Med Genet B Neuropsychiatr Genet 2016; 171:1032-1040. [PMID: 27312590 DOI: 10.1002/ajmg.b.32471] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/03/2016] [Indexed: 12/21/2022]
Abstract
Genome-wide association studies (GWASs) have identified multiple schizophrenia (SCZ) risk variants for samples of European and East Asian descent, but most of the identified susceptibility variants are population-specific to either Europeans or East Asians. This strong genetic heterogeneity suggests that differential population histories may play a role in SCZ susceptibility. Here, we explored this possibility by examining the allele frequency divergence of 136 previously reported genome-wide SCZ risk SNPs between European and East Asian populations. Our results showed that two SNPs (rs11038167 and rs11038172) at TSPAN18, reported as genome-wide significant SCZ risk variants in Han Chinese, were entirely monomorphic in Europeans, indicating a deep between-population divergence at this gene locus. To explore the evolutionary history of TSPAN18 in East Asians, we conducted population genetic analyses including multiple neutrality tests, the haplotype-based iHS and EHH tests, as well as haplotype bifurcation map and network constructions. We found that the protective allele of rs11038172 (G allele) had a long extended haplotype with much slower decay compared to the A allele. The star-like shape of the G-allele-carrying haplotypes indicates a recent enrichment in East Asians. Together, the evidences suggest that the protective allele of rs11038172 has experienced recent Darwinian positive selection in East Asians. These findings provide new insights that may help explain the strong genetic heterogeneity in SCZ risk and previous inconsistent association results for SCZ among both Europeans and East Asians. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jiewei Liu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| |
Collapse
|
64
|
Luykx JJ, Olde Loohuis LM, Neeleman M, Strengman E, Bakker SC, Lentjes E, Borgdorff P, van Dongen EPA, Bruins P, Kahn RS, Horvath S, de Jong S, Ophoff RA. Peripheral blood gene expression profiles linked to monoamine metabolite levels in cerebrospinal fluid. Transl Psychiatry 2016; 6:e983. [PMID: 27959337 PMCID: PMC5290339 DOI: 10.1038/tp.2016.245] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/15/2016] [Indexed: 01/07/2023] Open
Abstract
The blood-brain barrier separates circulating blood from the central nervous system (CNS). The scope of this barrier is not fully understood which limits our ability to relate biological measurements from peripheral to central phenotypes. For example, it is unknown to what extent gene expression levels in peripheral blood are reflective of CNS metabolism. In this study, we examine links between central monoamine metabolite levels and whole-blood gene expression to better understand the connection between peripheral systems and the CNS. To that end, we correlated the prime monoamine metabolites in cerebrospinal fluid (CSF) with whole-genome gene expression microarray data from blood (N=240 human subjects). We additionally applied gene-enrichment analysis and weighted gene co-expression network analyses (WGCNA) to identify modules of co-expressed genes in blood that may be involved with monoamine metabolite levels in CSF. Transcript levels of two genes were significantly associated with CSF serotonin metabolite levels after Bonferroni correction for multiple testing: THAP7 (P=2.8 × 10-8, β=0.08) and DDX6 (P=2.9 × 10-7, β=0.07). Differentially expressed genes were significantly enriched for genes expressed in the brain tissue (P=6.0 × 10-52). WGCNA revealed significant correlations between serotonin metabolism and hub genes with known functions in serotonin metabolism, for example, HTR2A and COMT. We conclude that gene expression levels in whole blood are associated with monoamine metabolite levels in the human CSF. Our results, including the strong enrichment of brain-expressed genes, illustrate that gene expression profiles in peripheral blood can be relevant for quantitative metabolic phenotypes in the CNS.
Collapse
Affiliation(s)
- J J Luykx
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands,Department of Translational Neuroscience Human Neurogenetics Unit, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands,Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium
| | - L M Olde Loohuis
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - M Neeleman
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - E Strengman
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S C Bakker
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - E Lentjes
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - P Borgdorff
- Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, The Netherlands
| | - E P A van Dongen
- Department of Anesthesiology, Intensive Care and Pain Management, University Medical Center Utrecht, Utrecht, The Netherlands
| | - P Bruins
- Department of Anesthesiology, Intensive Care and Pain Management, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R S Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA,Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
| | - S de Jong
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - R A Ophoff
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands,Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA,Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA,Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA. E-mail:
| |
Collapse
|
65
|
Minozzi G, Mattiello S, Grosso L, Crepaldi P, Chessa S, Pagnacco G. First insights in the genetics of caseous lymphadenitis in goats. ITALIAN JOURNAL OF ANIMAL SCIENCE 2016. [DOI: 10.1080/1828051x.2016.1250610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Giulietta Minozzi
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milano, Italy
| | - Silvana Mattiello
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milano, Italy
| | - Lilia Grosso
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milano, Italy
| | - Paola Crepaldi
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milano, Italy
| | - Stefania Chessa
- Consiglio Nazionale delle Ricerche, Istituto di Biologia e Biotecnologia Agraria, Lodi, Italy
| | - Giulio Pagnacco
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
66
|
Ozel MD, Onder ME, Sazci A. Transcription factor 4 gene rs9960767 polymorphism in bipolar disorder. Biomed Rep 2016; 5:506-510. [PMID: 27699022 DOI: 10.3892/br.2016.742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/08/2016] [Indexed: 12/18/2022] Open
Abstract
The transcription factor 4 (TCF4) gene encodes a helix-loop-helix transcription factor protein, which initiates neuronal differentiation and is primarily expressed during nervous system development. The aim of the present study is to investigate the association of the TCF4 rs9960767 polymorphism and bipolar disorder, which is highly heritable. DNA isolation was performed on 95 patients with bipolar disorder and 108 healthy control subjects to examine the TCF4 rs9960767 polymorphism. Genotypic and allelic frequencies were determined using the polymerase chain reaction-restriction fragment length polymorphism method designed in our laboratory. Statistical analysis was performed using χ2 test within the 95% confidence interval. Odds ratios were calculated and Hardy-Weinberg equilibrium (HWE) was verified for all control subjects and patients. The A allele frequency was 95.8% in the patients and 94.4% in the control subjects, and 4.2% in the patients and 5.6% in the control subjects for the C allele. The genotype frequencies of the TCF4 gene rs9960767 variant were as follows: AA, 91.6% and AC, 8.4% in patients with bipolar (CC genotype was not observed in cases); AA, 89.8%; AC, 9.3% and CC, 0.9% in the control subjects. No statistically significant difference was identified between the patients and control subjects (χ2=0.937; P=0.626). In addition, gender specific analysis was performed, although no significant association was found according to the gender distrubition. All patients and control subjects were in HWE (P>0.05). Statistical analysis of the data indicates that the TCF4 gene rs9960767 polymorphism is not an independent risk factor for bipolar disorder in the overall population or in terms of gender; however, an increased population size would improve the statistical power. Furthermore, additional gene variants that are specifically involved in neuronal development may be analyzed for revealing the complex genetic architecture of bipolar disorder. An improved approach would be better to evaluate the TCF4 gene in a pathway specific manner due to its role as a transcription factor.
Collapse
Affiliation(s)
- Mavi Deniz Ozel
- Department of Medical Biology, International School of Medicine, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Mehmet Emin Onder
- Department of Psychiatry, Faculty of Medicine, University of Kocaeli, Kocaeli 41380, Turkey
| | - Ali Sazci
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Kocaeli, Kocaeli 41380, Turkey
| |
Collapse
|
67
|
Tcf4 transgenic female mice display delayed adaptation in an auditory latent inhibition paradigm. Eur Arch Psychiatry Clin Neurosci 2016; 266:505-12. [PMID: 26404636 DOI: 10.1007/s00406-015-0643-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/07/2015] [Indexed: 12/18/2022]
Abstract
Schizophrenia (SZ) is a severe mental disorder affecting about 1 % of the human population. Patients show severe deficits in cognitive processing often characterized by an improper filtering of environmental stimuli. Independent genome-wide association studies confirmed a number of risk variants for SZ including several associated with the gene encoding the transcription factor 4 (TCF4). TCF4 is widely expressed in the central nervous system of mice and humans and seems to be important for brain development. Transgenic mice overexpressing murine Tcf4 (Tcf4tg) in the adult brain display cognitive impairments and sensorimotor gating disturbances. To address the question of whether increased Tcf4 gene dosage may affect cognitive flexibility in an auditory associative task, we tested latent inhibition (LI) in female Tcf4tg mice. LI is a widely accepted translational endophenotype of SZ and results from a maladaptive delay in switching a response to a previously unconditioned stimulus when this becomes conditioned. Using an Audiobox, we pre-exposed Tcf4tg mice and their wild-type littermates to either a 3- or a 12-kHz tone before conditioning them to a 12-kHz tone. Tcf4tg animals pre-exposed to a 12-kHz tone showed significantly delayed conditioning when the previously unconditioned tone became associated with an air puff. These results support findings that associate TCF4 dysfunction with cognitive inflexibility and improper filtering of sensory stimuli observed in SZ patients.
Collapse
|
68
|
Need AC. Neuropsychiatric genomics in precision medicine: diagnostics, gene discovery, and translation. DIALOGUES IN CLINICAL NEUROSCIENCE 2016; 18:237-252. [PMID: 27757059 PMCID: PMC5067142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Only a few years after its development, next-generation sequencing is rapidly becoming an essential part of clinical care for patients with serious neurological conditions, especially in the diagnosis of early-onset and severe presentations. Beyond this diagnostic role, there has been an explosion in definitive gene discovery in a range of neuropsychiatric diseases. This is providing new pointers to underlying disease biology and is beginning to outline a new framework for genetic stratification of neuropsychiatric disease, with clear relevance to both individual treatment optimization and clinical trial design. Here, we outline these developments and chart the expected impact on the treatment of neurological, neurodevelopmental, and psychiatric disease.
Collapse
Affiliation(s)
- Anna C. Need
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, W12 ONN, UK
| |
Collapse
|
69
|
Chang H, Zhang C, Xiao X, Pu X, Liu Z, Wu L, Li M. Further evidence of VRK2 rs2312147 associated with schizophrenia. World J Biol Psychiatry 2016; 17:457-66. [PMID: 27382989 DOI: 10.1080/15622975.2016.1200746] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVES Previous genome-wide association studies (GWAS) have reported that rs2312147 near the VRK2 gene was significantly associated with schizophrenia in populations of European descent, but negative results have also been observed. METHODS To perform a systematic meta-analysis, we collected statistical data of rs2312147 from both GWAS and individual replication samples in European and Asian populations, which finally included up to 30,867 schizophrenia patients and 59,863 healthy controls. RESULTS The VRK2 rs2312147 was genome-wide significantly associated with schizophrenia in combined populations (P = 1.31 × 10(-15), odds ratio, OR = 1.10) as well as in Europeans only (P = 2.35 × 10(-12), OR =1.09). In Asian samples, the SNP did not reach genome-wide level of statistical significance (P = 1.23 × 10 (-) (5), OR =1.19), which is likely due to the limited power of small sample size in this population (2,974 cases and 4,786 controls). However, the effect size of rs2312147 did not alter significantly between populations, and is also in agreement with the observed effect sizes of other genetic risk loci in large scale studies. CONCLUSIONS Our data provides further evidence for the genetic contributions of VRK2 rs2312147 to schizophrenia susceptibility especially in Europeans, while further replication analyses in Asian populations are still needed, and future studies, e.g., the underlying molecular mechanisms of genetic risk, are necessary.
Collapse
Affiliation(s)
- Hong Chang
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province , Kunming Institute of Zoology , Kunming , Yunnan , China
| | - Chen Zhang
- b Division of Mood Disorders , Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Xiao Xiao
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province , Kunming Institute of Zoology , Kunming , Yunnan , China
| | - Xingfu Pu
- c The Second People's Hospital of Yuxi City , Yuxi , Yunnan , China
| | - Zichao Liu
- d Key Laboratory of Special Biological Resource Development and Utilization of Universities in Yunnan Province, Department of Biological Science and Technology , Kunming University , Kunming , Yunnan , China
| | - Lichuan Wu
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province , Kunming Institute of Zoology , Kunming , Yunnan , China
| | - Ming Li
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province , Kunming Institute of Zoology , Kunming , Yunnan , China
| |
Collapse
|
70
|
Tesli M, Wirgenes KV, Hughes T, Bettella F, Athanasiu L, Hoseth ES, Nerhus M, Lagerberg TV, Steen NE, Agartz I, Melle I, Dieset I, Djurovic S, Andreassen OA. VRK2 gene expression in schizophrenia, bipolar disorder and healthy controls. Br J Psychiatry 2016; 209:114-20. [PMID: 26941264 DOI: 10.1192/bjp.bp.115.161950] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 09/24/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Common variants in the Vaccinia-related kinase 2 (VRK2) gene have been associated with schizophrenia, but the relevance of its encoded protein VRK2 in the disorder remains unclear. AIMS To identify potential differences in VRK2 gene expression levels between schizophrenia, bipolar disorder, psychosis not otherwise specified (PNOS) and healthy controls. METHOD VRK2 mRNA level was measured in whole blood in 652 individuals (schizophrenia, n = 201; bipolar disorder, n = 167; PNOS, n = 61; healthy controls, n = 223), and compared across diagnostic categories and subcategories. Additionally, we analysed for association between 1566 VRK2 single nucleotide polymorphisms and mRNA levels. RESULTS We found lower VRK2 mRNA levels in schizophrenia compared with healthy controls (P<10(-12)), bipolar disorder (P<10(-12)) and PNOS (P = 0.0011), and lower levels in PNOS than in healthy controls (P = 0.0042) and bipolar disorder (P = 0.00026). Expression quantitative trait loci in close proximity to the transcription start site of the short isoforms of the VRK2 gene were identified. CONCLUSIONS Altered VRK2 gene expression seems specific for schizophrenia and PNOS, which is in accordance with findings from genome-wide association studies. These results suggest that reduced VRK2 mRNA levels are involved in the underlying mechanisms in schizophrenia spectrum disorders.
Collapse
Affiliation(s)
- Martin Tesli
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Katrine Verena Wirgenes
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Timothy Hughes
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Francesco Bettella
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Lavinia Athanasiu
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Eva S Hoseth
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Mari Nerhus
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Trine V Lagerberg
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Nils E Steen
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Ingrid Agartz
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Ingrid Melle
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Ingrid Dieset
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Srdjan Djurovic
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| | - Ole A Andreassen
- Martin Tesli, MD, PhD, Katrine Verena Wirgenes, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Timothy Hughes, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Francesco Bettella, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Lavinia Athanasiu, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Eva S. Hoseth, MD, Mari Nerhus, MD, Trine V. Lagerberg, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Nils E. Steen, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Drammen District Psychiatric Centre, Clinic of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway; Ingrid Agartz, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Ingrid Melle, MD, PhD, Ingrid Dieset, MD, PhD, NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, Univ
| |
Collapse
|
71
|
Glycogen synthase kinase 3β suppresses polyglutamine aggregation by inhibiting Vaccinia-related kinase 2 activity. Sci Rep 2016; 6:29097. [PMID: 27377031 PMCID: PMC4932512 DOI: 10.1038/srep29097] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/09/2016] [Indexed: 12/19/2022] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by an abnormal expansion of polyglutamine repeats in the N-terminal of huntingtin. The amount of aggregate-prone protein is controlled by various mechanisms, including molecular chaperones. Vaccinia-related kinase 2 (VRK2) is known to negatively regulate chaperonin TRiC, and VRK2-facilitated degradation of TRiC increases polyQ protein aggregation, which is involved in HD. We found that VRK2 activity was negatively controlled by glycogen synthase kinase 3β (GSK3β). GSK3β directly bound to VRK2 and inhibited the catalytic activity of VRK2 in a kinase activity-independent manner. Furthermore, GSK3β increased the stability of TRiC and decreased the formation of HttQ103-GFP aggregates by inhibiting VRK2. These results indicate that GSK3β signaling may be a regulatory mechanism of HD progression and suggest targets for further therapeutic trials for HD.
Collapse
|
72
|
González-Peñas J, Amigo J, Santomé L, Sobrino B, Brenlla J, Agra S, Paz E, Páramo M, Carracedo Á, Arrojo M, Costas J. Targeted resequencing of regulatory regions at schizophrenia risk loci: Role of rare functional variants at chromatin repressive states. Schizophr Res 2016; 174:10-16. [PMID: 27066855 DOI: 10.1016/j.schres.2016.03.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 03/24/2016] [Indexed: 12/30/2022]
Abstract
There is mounting evidence that regulatory variation plays an important role in genetic risk for schizophrenia. Here, we specifically search for regulatory variants at risk by sequencing promoter regions of twenty-three genes implied in schizophrenia by copy number variant or genome-wide association studies. After strict quality control, a total of 55,206bp per sample were analyzed in 526 schizophrenia cases and 516 controls from Galicia, NW Spain, using the Applied Biosystems SOLiD System. Variants were filtered based on frequency from public databases, chromatin states from the RoadMap Epigenomics Consortium at tissues relevant for schizophrenia, such as fetal brain, mid-frontal lobe, and angular gyrus, and prediction of functionality from RegulomeDB. The proportion of rare variants at polycomb repressive chromatin state at relevant tissues was higher in cases than in controls. The proportion of rare variants with predicted regulatory role was significantly higher in cases than in controls (P=0.0028, OR=1.93, 95% C.I.=1.23-3.04). Combination of information from both sources led to the identification of an excess of carriers of rare variants with predicted regulatory role located at polycomb repressive chromatin state at relevant tissues in cases versus controls (P=0.0016, OR=19.34, 95% C.I.=2.45-2495.26). The variants are located at two genes affected by the 17q12 copy number variant, LHX1 and HNF1B. These data strongly suggest that a specific epigenetic mechanism, chromatin remodeling by histone modification during early development, may be impaired in a subset of schizophrenia patients, in agreement with previous data.
Collapse
Affiliation(s)
- Javier González-Peñas
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Jorge Amigo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Fundación Pública Galega de Medicina Xenómica, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Luis Santomé
- Fundación Pública Galega de Medicina Xenómica, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Beatriz Sobrino
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Fundación Pública Galega de Medicina Xenómica, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Julio Brenlla
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Santiago Agra
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Eduardo Paz
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Mario Páramo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Ángel Carracedo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Fundación Pública Galega de Medicina Xenómica, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Manuel Arrojo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Javier Costas
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain.
| |
Collapse
|
73
|
Xu Y, Yue W, Yao Shugart Y, Li S, Cai L, Li Q, Cheng Z, Wang G, Zhou Z, Jin C, Yuan J, Tian L, Wang J, Zhang K, Zhang K, Liu S, Song Y, Zhang F. Exploring Transcription Factors-microRNAs Co-regulation Networks in Schizophrenia. Schizophr Bull 2016; 42:1037-45. [PMID: 26609121 PMCID: PMC4903044 DOI: 10.1093/schbul/sbv170] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Transcriptional factors (TFs) and microRNAs (miRNAs) have been recognized as 2 classes of principal gene regulators that may be responsible for genome coexpression changes observed in schizophrenia (SZ). METHODS This study aims to (1) identify differentially coexpressed genes (DCGs) in 3 mRNA expression microarray datasets; (2) explore potential interactions among the DCGs, and differentially expressed miRNAs identified in our dataset composed of early-onset SZ patients and healthy controls; (3) validate expression levels of some key transcripts; and (4) explore the druggability of DCGs using the curated database. RESULTS We detected a differential coexpression network associated with SZ and found that 9 out of the 12 regulators were replicated in either of the 2 other datasets. Leveraging the differentially expressed miRNAs identified in our previous dataset, we constructed a miRNA-TF-gene network relevant to SZ, including an EGR1-miR-124-3p-SKIL feed-forward loop. Our real-time quantitative PCR analysis indicated the overexpression of miR-124-3p, the under expression of SKIL and EGR1 in the blood of SZ patients compared with controls, and the direction of change of miR-124-3p and SKIL mRNA levels in SZ cases were reversed after a 12-week treatment cycle. Our druggability analysis revealed that many of these genes have the potential to be drug targets. CONCLUSIONS Together, our results suggest that coexpression network abnormalities driven by combinatorial and interactive action from TFs and miRNAs may contribute to the development of SZ and be relevant to the clinical treatment of the disease.
Collapse
Affiliation(s)
- Yong Xu
- Department of Psychiatry, First Clinical Medical College/First Hospital of Shanxi Medical University, Taiyuan, China;,These authors contributed equally to this work
| | - Weihua Yue
- Department of Psychiatry, Institute of Mental Health, Sixth Hospital, Peking University, Beijing, China;,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), Beijing, China;,These authors contributed equally to this work
| | - Yin Yao Shugart
- Unit on Statistical Genomics, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD;,These authors contributed equally to this work
| | - Sheng Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Cai
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Li
- Shanghai Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, China
| | - Zaohuo Cheng
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Guoqiang Wang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Zhenhe Zhou
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Chunhui Jin
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Jianmin Yuan
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Lin Tian
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Jun Wang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Kai Zhang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Kerang Zhang
- Department of Psychiatry, First Clinical Medical College/First Hospital of Shanxi Medical University, Taiyuan, China
| | - Sha Liu
- Department of Psychiatry, First Clinical Medical College/First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuqing Song
- Department of Psychiatry, Institute of Mental Health, Sixth Hospital, Peking University, Beijing, China
| | - Fuquan Zhang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| |
Collapse
|
74
|
Xiao X, Luo XJ, Chang H, Liu Z, Li M. Evaluation of European Schizophrenia GWAS Loci in Asian Populations via Comprehensive Meta-Analyses. Mol Neurobiol 2016; 54:4071-4080. [DOI: 10.1007/s12035-016-9990-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 06/14/2016] [Indexed: 11/30/2022]
|
75
|
D'Rozario M, Zhang T, Waddell EA, Zhang Y, Sahin C, Sharoni M, Hu T, Nayal M, Kutty K, Liebl F, Hu W, Marenda DR. Type I bHLH Proteins Daughterless and Tcf4 Restrict Neurite Branching and Synapse Formation by Repressing Neurexin in Postmitotic Neurons. Cell Rep 2016; 15:386-97. [PMID: 27050508 DOI: 10.1016/j.celrep.2016.03.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 01/22/2016] [Accepted: 03/09/2016] [Indexed: 11/17/2022] Open
Abstract
Proneural proteins of the class I/II basic-helix-loop-helix (bHLH) family are highly conserved transcription factors. Class I bHLH proteins are expressed in a broad number of tissues during development, whereas class II bHLH protein expression is more tissue restricted. Our understanding of the function of class I/II bHLH transcription factors in both invertebrate and vertebrate neurobiology is largely focused on their function as regulators of neurogenesis. Here, we show that the class I bHLH proteins Daughterless and Tcf4 are expressed in postmitotic neurons in Drosophila melanogaster and mice, respectively, where they function to restrict neurite branching and synapse formation. Our data indicate that Daughterless performs this function in part by restricting the expression of the cell adhesion molecule Neurexin. This suggests a role for these proteins outside of their established roles in neurogenesis.
Collapse
Affiliation(s)
| | - Ting Zhang
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Edward A Waddell
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Yonggang Zhang
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Cem Sahin
- Department of Electrical and Computer Engineering, Drexel University, Philadelphia, PA 19104, USA
| | - Michal Sharoni
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Tina Hu
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Mohammad Nayal
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Kaveesh Kutty
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Faith Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL 62026, USA
| | - Wenhui Hu
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| | - Daniel R Marenda
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
76
|
Moons T, De Hert M, Gellens E, Gielen L, Sweers K, Jacqmaert S, van Winkel R, Vandekerckhove P, Claes S. Genetic Evaluation of Schizophrenia Using the Illumina HumanExome Chip. PLoS One 2016; 11:e0150464. [PMID: 27028512 PMCID: PMC4814136 DOI: 10.1371/journal.pone.0150464] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 02/15/2016] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Schizophrenia is a genetically heterogeneous disorder that is associated with several common and rare genetic variants. As technology involved, cost advantages of chip based genotyping was combined with information about rare variants, resulting in the Infinium HumanExome Beadchip. Using this chip, a sample of 493 patients with schizophrenia or schizoaffective disorder and 484 healthy controls was genotyped. RESULTS From the initial 242901 SNVs, 88306 had at least one minor allele and passed quality control. No variant reached genomewide-significant results (p<10(-8)). The SNP with the lowest p-value was rs1230345 in WISP3 (p = 3.05*10(-6)), followed by rs9311525 in CACNA2D3 (p = 1.03*10(-5)) and rs1558557 (p = 3.85*10(-05)) on chromosome 7. At the gene level, 3 genes were of interest: WISP3, on chromosome 6q21, a signally protein from the extracellular matrix. A second candidate gene is CACNA2D3, a regulator of the intracerebral calcium pathway. A third gene is TNFSF10, associated with p53 mediated apoptosis.
Collapse
Affiliation(s)
- Tim Moons
- GRASP research group, UPC KULeuven, Campus Leuven, Leuven, Belgium
| | - Marc De Hert
- UPC KULeuven, campus Kortenberg, Kortenberg, Belgium
| | - Edith Gellens
- GRASP research group, UPC KULeuven, Campus Leuven, Leuven, Belgium
| | - Leen Gielen
- UPC KULeuven, campus Kortenberg, Kortenberg, Belgium
| | - Kim Sweers
- UPC KULeuven, campus Kortenberg, Kortenberg, Belgium
| | | | - Ruud van Winkel
- KU Leuven—University of Leuven, Department of Public Health and Primary Care, Leuven, Belgium
| | - Philippe Vandekerckhove
- Belgian Red Cross-Flanders, Mechelen, Belgium
- KU Leuven—University of Leuven, Department of Public Health and Primary Care, Leuven, Belgium
| | - Stephan Claes
- GRASP research group, UPC KULeuven, Campus Leuven, Leuven, Belgium
| |
Collapse
|
77
|
Costas J, Carrera N, Alonso P, Gurriarán X, Segalàs C, Real E, López-Solà C, Mas S, Gassó P, Domènech L, Morell M, Quintela I, Lázaro L, Menchón JM, Estivill X, Carracedo Á. Exon-focused genome-wide association study of obsessive-compulsive disorder and shared polygenic risk with schizophrenia. Transl Psychiatry 2016; 6:e768. [PMID: 27023174 PMCID: PMC4872458 DOI: 10.1038/tp.2016.34] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/11/2015] [Accepted: 02/10/2016] [Indexed: 12/17/2022] Open
Abstract
Common single-nucleotide polymorphisms (SNPs) account for a large proportion of the heritability of obsessive-compulsive disorder (OCD). Co-ocurrence of OCD and schizophrenia is commoner than expected based on their respective prevalences, complicating the clinical management of patients. This study addresses two main objectives: to identify particular genes associated with OCD by SNP-based and gene-based tests; and to test the existence of a polygenic risk shared with schizophrenia. The primary analysis was an exon-focused genome-wide association study of 370 OCD cases and 443 controls from Spain. A polygenic risk model based on the Psychiatric Genetics Consortium schizophrenia data set (PGC-SCZ2) was tested in our OCD data. A polygenic risk model based on our OCD data was tested on previous data of schizophrenia from our group. The most significant association at the gene-based test was found at DNM3 (P=7.9 × 10(-5)), a gene involved in synaptic vesicle endocytosis. The polygenic risk model from PGC-SCZ2 data was strongly associated with disease status in our OCD sample, reaching its most significant value after removal of the major histocompatibility complex region (lowest P=2.3 × 10(-6), explaining 3.7% of the variance). The shared polygenic risk was confirmed in our schizophrenia data. In conclusion, DNM3 may be involved in risk to OCD. The shared polygenic risk between schizophrenia and OCD may be partially responsible for the frequent comorbidity of both disorders, explaining epidemiological data on cross-disorder risk. This common etiology may have clinical implications.
Collapse
Affiliation(s)
- J Costas
- Grupo de Xenética Psiquiátrica, Instituto de Investigación Sanitaria de Santiago, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde, Santiago de Compostela, Spain,Grupo de Xenética Psiquiátrica, Instituto de Investigación Sanitaria de Santiago, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde, despacho 15, E-15706 Santiago de Compostela, Spain. E-mail:
| | - N Carrera
- Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK,Fundación Pública Galega de Medicina Xenómica, Servicio Galego de Saúde, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - P Alonso
- OCD Clinical and Research Unit, Department of Psychiatry, Hospital de Bellvitge, Barcelona, Spain,Bellvitge Biomedical Research Institute-IDIBELL, Barcelona, Spain,Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain,Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain
| | - X Gurriarán
- Grupo de Xenética Psiquiátrica, Instituto de Investigación Sanitaria de Santiago, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde, Santiago de Compostela, Spain,Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - C Segalàs
- OCD Clinical and Research Unit, Department of Psychiatry, Hospital de Bellvitge, Barcelona, Spain,Bellvitge Biomedical Research Institute-IDIBELL, Barcelona, Spain,Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain
| | - E Real
- OCD Clinical and Research Unit, Department of Psychiatry, Hospital de Bellvitge, Barcelona, Spain,Bellvitge Biomedical Research Institute-IDIBELL, Barcelona, Spain,Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain
| | - C López-Solà
- OCD Clinical and Research Unit, Department of Psychiatry, Hospital de Bellvitge, Barcelona, Spain,Bellvitge Biomedical Research Institute-IDIBELL, Barcelona, Spain,Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain
| | - S Mas
- Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain,Department of Anatomic Pathology, Pharmacology and Microbiology, University of Barcelona, Barcelona, Spain,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - P Gassó
- Department of Anatomic Pathology, Pharmacology and Microbiology, University of Barcelona, Barcelona, Spain,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain,Department of Psychiatry and Clinical Psychobiology, University of Barcelona, Barcelona, Spain
| | - L Domènech
- Genomics and Disease Group, Centre for Genomic Regulation, Barcelona, Spain,Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain,Centro de Investigación Biomédica en Red Epidemiología y Salud Pública, Centre for Genomic Regulation, Barcelona, Spain,Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - M Morell
- Genomics and Disease Group, Centre for Genomic Regulation, Barcelona, Spain,Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain,Centro de Investigación Biomédica en Red Epidemiología y Salud Pública, Centre for Genomic Regulation, Barcelona, Spain,Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - I Quintela
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro Nacional de Genotipado - Instituto Carlos III, Santiago de Compostela, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras, Santiago de Compostela, Spain
| | - L Lázaro
- Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain,Department of Psychiatry and Clinical Psychobiology, University of Barcelona, Barcelona, Spain,Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clínic Universitari, Barcelona, Spain
| | - J M Menchón
- OCD Clinical and Research Unit, Department of Psychiatry, Hospital de Bellvitge, Barcelona, Spain,Bellvitge Biomedical Research Institute-IDIBELL, Barcelona, Spain,Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain,Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain
| | - X Estivill
- Genomics and Disease Group, Centre for Genomic Regulation, Barcelona, Spain,Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain,Centro de Investigación Biomédica en Red Epidemiología y Salud Pública, Centre for Genomic Regulation, Barcelona, Spain,Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Á Carracedo
- Fundación Pública Galega de Medicina Xenómica, Servicio Galego de Saúde, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain,Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro Nacional de Genotipado - Instituto Carlos III, Santiago de Compostela, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras, Santiago de Compostela, Spain
| |
Collapse
|
78
|
Li M, Luo XJ, Landén M, Bergen SE, Hultman CM, Li X, Zhang W, Yao YG, Zhang C, Liu J, Mattheisen M, Cichon S, Mühleisen TW, Degenhardt FA, Nöthen MM, Schulze TG, Grigoroiu-Serbanescu M, Li H, Fuller CK, Chen C, Dong Q, Chen C, Jamain S, Leboyer M, Bellivier F, Etain B, Kahn JP, Henry C, Preisig M, Kutalik Z, Castelao E, Wright A, Mitchell PB, Fullerton JM, Schofield PR, Montgomery GW, Medland SE, Gordon SD, Martin NG, Rietschel M, Liu C, Kleinman JE, Hyde TM, Weinberger DR, Su B. Impact of a cis-associated gene expression SNP on chromosome 20q11.22 on bipolar disorder susceptibility, hippocampal structure and cognitive performance. Br J Psychiatry 2016; 208:128-137. [PMID: 26338991 PMCID: PMC4829352 DOI: 10.1192/bjp.bp.114.156976] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 10/21/2014] [Indexed: 11/23/2022]
Abstract
BACKGROUND Bipolar disorder is a highly heritable polygenic disorder. Recent enrichment analyses suggest that there may be true risk variants for bipolar disorder in the expression quantitative trait loci (eQTL) in the brain. AIMS We sought to assess the impact of eQTL variants on bipolar disorder risk by combining data from both bipolar disorder genome-wide association studies (GWAS) and brain eQTL. METHOD To detect single nucleotide polymorphisms (SNPs) that influence expression levels of genes associated with bipolar disorder, we jointly analysed data from a bipolar disorder GWAS (7481 cases and 9250 controls) and a genome-wide brain (cortical) eQTL (193 healthy controls) using a Bayesian statistical method, with independent follow-up replications. The identified risk SNP was then further tested for association with hippocampal volume (n = 5775) and cognitive performance (n = 342) among healthy individuals. RESULTS Integrative analysis revealed a significant association between a brain eQTL rs6088662 on chromosome 20q11.22 and bipolar disorder (log Bayes factor = 5.48; bipolar disorder P = 5.85 × 10(-5)). Follow-up studies across multiple independent samples confirmed the association of the risk SNP (rs6088662) with gene expression and bipolar disorder susceptibility (P = 3.54 × 10(-8)). Further exploratory analysis revealed that rs6088662 is also associated with hippocampal volume and cognitive performance in healthy individuals. CONCLUSIONS Our findings suggest that 20q11.22 is likely a risk region for bipolar disorder; they also highlight the informative value of integrating functional annotation of genetic variants for gene expression in advancing our understanding of the biological basis underlying complex disorders, such as bipolar disorder.
Collapse
Affiliation(s)
- Ming Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Lieber Institute for Brain Development, Johns Hopkins University, Baltimore, Maryland, USA
| | - Xiong-jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- University of Rochester Flaum Eye Institute, University of Rochester, Rochester, New York, USA
| | - Mikael Landén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Section of Psychiatry and Neurochemistry, Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| | - Sarah E. Bergen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Christina M. Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Xiao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Wen Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Chen Zhang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiewei Liu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | | | - Sven Cichon
- Division of Medical Genetics, University of Basel, Basel, Switzerland
- Institute of Human Genetics and Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Structural and Functional Organization of the Brain, Genomic Imaging, Research Centre Jülich, D-52425 Jülich, Germany
| | - Thomas W. Mühleisen
- Institute of Human Genetics and Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Structural and Functional Organization of the Brain, Genomic Imaging, Research Centre Jülich, D-52425 Jülich, Germany
| | - Franziska A. Degenhardt
- Institute of Human Genetics and Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Markus M. Nöthen
- Institute of Human Genetics and Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Thomas G. Schulze
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/University of Heidelberg, Mannheim, Germany
- Section on Psychiatric Genetics, Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Maria Grigoroiu-Serbanescu
- Biometric Psychiatric Genetics Research Unit, Alexandru Obregia Clinical Psychiatric Hospital, Bucharest, Romania
| | - Hao Li
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California, USA
| | - Chris K. Fuller
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California, USA
| | - Chunhui Chen
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Center for Collaboration and Innovation in Brain and Learning Sciences, Beijing Normal University, Beijing, China
| | - Qi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Center for Collaboration and Innovation in Brain and Learning Sciences, Beijing Normal University, Beijing, China
| | - Chuansheng Chen
- Department of Psychology and Social Behavior, University of California, Irvine, California, USA
| | - Stéphane Jamain
- Inserm U 955, IMRB, Psychiatrie Génétique, Créteil, France
- Université Paris Est, Faculté de Médecine, Créteil, France
- Fondation Fondamental, Créteil, France
| | - Marion Leboyer
- Inserm U 955, IMRB, Psychiatrie Génétique, Créteil, France
- Université Paris Est, Faculté de Médecine, Créteil, France
- Fondation Fondamental, Créteil, France
- AP-HP, Hôpital A. Chenevier - H. Mondor, Pôle de Psychiatrie, Créteil, France
| | - Frank Bellivier
- Inserm U 955, IMRB, Psychiatrie Génétique, Créteil, France
- Fondation Fondamental, Créteil, France
- AP-HP, Groupe hospitalier Lariboisière - F. Widal, Pôle de Psychiatrie, Paris, France
- Université Paris Diderot, Paris, France
| | - Bruno Etain
- Inserm U 955, IMRB, Psychiatrie Génétique, Créteil, France
- Université Paris Est, Faculté de Médecine, Créteil, France
- Fondation Fondamental, Créteil, France
- AP-HP, Hôpital A. Chenevier - H. Mondor, Pôle de Psychiatrie, Créteil, France
| | - Jean-Pierre Kahn
- Fondation Fondamental, Créteil, France
- Département de Psychiatrie et de Psychologie Clinique, CHU de Nancy, Hôpital Jeanne d'Arc, Toul, France
| | - Chantal Henry
- Inserm U 955, IMRB, Psychiatrie Génétique, Créteil, France
- Université Paris Est, Faculté de Médecine, Créteil, France
- Fondation Fondamental, Créteil, France
- AP-HP, Hôpital A. Chenevier - H. Mondor, Pôle de Psychiatrie, Créteil, France
| | - Martin Preisig
- Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, Prilly, Switzerland
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Enrique Castelao
- Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, Prilly, Switzerland
| | - Adam Wright
- School of Psychiatry, University of New South Wales, Randwick, Australia
- Black Dog Institute, Prince of Wales Hospital, Randwick, Australia
| | - Philip B. Mitchell
- School of Psychiatry, University of New South Wales, Randwick, Australia
- Black Dog Institute, Prince of Wales Hospital, Randwick, Australia
| | - Janice M. Fullerton
- Neuroscience Research Australia, Randwick, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Peter R. Schofield
- Neuroscience Research Australia, Randwick, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | | | | | - Scott D. Gordon
- Queensland Institute of Medical Research, Brisbane, Australia
| | | | | | | | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/University of Heidelberg, Mannheim, Germany
| | - Chunyu Liu
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, Johns Hopkins University, Baltimore, Maryland, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel R. Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| |
Collapse
|
79
|
Ahn K, An SS, Shugart YY, Rapoport JL. Common polygenic variation and risk for childhood-onset schizophrenia. Mol Psychiatry 2016; 21:94-6. [PMID: 25510512 DOI: 10.1038/mp.2014.158] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/02/2014] [Accepted: 10/09/2014] [Indexed: 12/16/2022]
Abstract
Childhood-onset schizophrenia (COS) is a rare and severe form of the disorder, with more striking abnormalities with respect to prepsychotic developmental disorders and abnormities in the brain development compared with later-onset schizophrenia. We previously documented that COS patients, compared with their healthy siblings and with adult-onset patients (AOS), carry significantly more rare chromosomal copy number variations, spanning large genomic regions (>100 kb) (Ahn et al. 2014). Here, we interrogated the contribution of common polygenic variation to the genetic susceptibility for schizophrenia. We examined the association between a direct measure of genetic risk of schizophrenia in 130 COS probands and 103 healthy siblings. Using data from the schizophrenia and autism GWAS of the Psychiatric Genomic Consortia, we selected three risk-related sets of single nucleotide polymorphisms from which we conducted polygenic risk score comparisons for COS probands and their healthy siblings. COS probands had higher genetic risk scores of both schizophrenia and autism than their siblings (P<0.05). Given the small sample size, these findings suggest that COS patients have more salient genetic risk than do AOS.
Collapse
Affiliation(s)
- K Ahn
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - S S An
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Y Y Shugart
- Unit of Statistical Genomics, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - J L Rapoport
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
80
|
Hu X, Fan Q, Hou H, Yan R. Neurological dysfunctions associated with altered BACE1-dependent Neuregulin-1 signaling. J Neurochem 2016; 136:234-49. [PMID: 26465092 PMCID: PMC4833723 DOI: 10.1111/jnc.13395] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 01/09/2023]
Abstract
Inhibition of BACE1 is being pursued as a therapeutic target to treat patients suffering from Alzheimer's disease because BACE1 is the sole β-secretase that generates β-amyloid peptide. Knowledge regarding other cellular functions of BACE1 is therefore critical for the safe use of BACE1 inhibitors in human patients. Neuregulin-1 (Nrg1) is a BACE1 substrate and BACE1 cleavage of Nrg1 is critical for signaling functions in myelination, remyelination, synaptic plasticity, normal psychiatric behaviors, and maintenance of muscle spindles. This review summarizes the most recent discoveries associated with BACE1-dependent Nrg1 signaling in these areas. This body of knowledge will help to provide guidance for preventing unwanted Nrg1-based side effects following BACE1 inhibition in humans. To initiate its signaling cascade, membrane anchored Neuregulin (Nrg), mainly type I and III β1 Nrg1 isoforms and Nrg3, requires ectodomain shedding. BACE1 is one of such indispensable sheddases to release the functional Nrg signaling fragment. The dependence of Nrg on the cleavage by BACE1 is best manifested by disrupting the critical role of Nrg in the control of axonal myelination, schizophrenic behaviors as well as the formation and maintenance of muscle spindles.
Collapse
Affiliation(s)
- Xiangyou Hu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Qingyuan Fan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Hailong Hou
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Riqiang Yan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
81
|
McKay GJ, Kavanagh DH, Crean JK, Maxwell AP. Bioinformatic Evaluation of Transcriptional Regulation of WNT Pathway Genes with reference to Diabetic Nephropathy. J Diabetes Res 2016; 2016:7684038. [PMID: 26697505 PMCID: PMC4677197 DOI: 10.1155/2016/7684038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 05/18/2015] [Accepted: 05/24/2015] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE WNT/β-catenin pathway members have been implicated in interstitial fibrosis and glomerular sclerosis disease processes characteristic of diabetic nephropathy (DN), processes partly controlled by transcription factors (TFs) that bind to gene promoter regions attenuating regulation. We sought to identify predicted cis-acting transcription factor binding sites (TFBSs) overrepresented within WNT pathway members. METHODS We assessed 62 TFBS motif frequencies from the JASPAR databases in 65 WNT pathway genes. P values were estimated on the hypergeometric distribution for each TF. Gene expression profiles of enriched motifs were examined in DN-related datasets to assess clinical significance. RESULTS Transcription factor AP-2 alpha (TFAP2A), myeloid zinc finger 1 (MZF1), and specificity protein 1 (SP1) were significantly enriched within WNT pathway genes (P values < 6.83 × 10(-29), 1.34 × 10(-11), and 3.01 × 10(-6), resp.). MZF1 expression was significantly increased in DN in a whole kidney dataset (fold change = 1.16; 16% increase; P = 0.03). TFAP2A expression was decreased in an independent dataset (fold change = -1.02; P = 0.03). No differential expression of SP1 was detected. CONCLUSIONS Three TFBS profiles are significantly enriched within WNT pathway genes highlighting the potential of in silico analyses for identification of pathway regulators. Modification of TF binding may possibly limit DN progression, offering potential therapeutic benefit.
Collapse
Affiliation(s)
- Gareth J. McKay
- Centre for Public Health, Queen's University Belfast, Belfast BT12 6BA, UK
- *Gareth J. McKay:
| | - David H. Kavanagh
- Centre for Public Health, Queen's University Belfast, Belfast BT12 6BA, UK
| | - John K. Crean
- Conway Institute, University College Dublin, Dublin 4, Ireland
| | | |
Collapse
|
82
|
González-Peñas J, Arrojo M, Paz E, Brenlla J, Páramo M, Costas J. Cumulative role of rare and common putative functional genetic variants at NPAS3 in schizophrenia susceptibility. Am J Med Genet B Neuropsychiatr Genet 2015; 168:528-35. [PMID: 25982957 DOI: 10.1002/ajmg.b.32324] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 05/04/2015] [Indexed: 11/07/2022]
Abstract
Schizophrenia may be considered a human-specific disorder arisen as a maladaptive by-product of human-specific brain evolution. Therefore, genetic variants involved in susceptibility to schizophrenia may be identified among those genes related to acquisition of human-specific traits. NPAS3, a transcription factor involved in central nervous system development and neurogenesis, seems to be implicated in the evolution of human brain, as it is the human gene with most human-specific accelerated elements (HAEs), i.e., .mammalian conserved regulatory sequences with accelerated evolution in the lineage leading to humans after human-chimpanzee split. We hypothesize that any nucleotide variant at the NPAS3 HAEs may lead to altered susceptibility to schizophrenia. Twenty-one variants at these HAEs detected by the 1000 genomes Project, as well as five additional variants taken from psychiatric genome-wide association studies, were genotyped in 538 schizophrenic patients and 539 controls from Galicia. Analyses at the haplotype level or based on the cumulative role of the variants assuming different susceptibility models did not find any significant association in spite of enough power under several plausible scenarios regarding direction of effect and the specific role of rare and common variants. These results suggest that, contrary to our hypothesis, the special evolution of the NPAS3 HAEs in Homo relaxed the strong constraint on sequence that characterized these regions during mammalian evolution, allowing some sequence changes without any effect on schizophrenia risk.
Collapse
Affiliation(s)
- Javier González-Peñas
- Instituto de investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain.,Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Arrojo
- Instituto de investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain.,Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Eduardo Paz
- Instituto de investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain.,Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Julio Brenlla
- Instituto de investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain.,Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Mario Páramo
- Instituto de investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain.,Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Javier Costas
- Instituto de investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| |
Collapse
|
83
|
Hui L, Rao WW, Yu Q, Kou C, Wu JQ, He JC, Ye MJ, Liu JH, Xu XJ, Zheng K, Ruan LN, Liu HY, Hu WM, Shao TN, AngRabanes MB, Soares JC, Zhang XY. TCF4 gene polymorphism is associated with cognition in patients with schizophrenia and healthy controls. J Psychiatr Res 2015; 69:95-101. [PMID: 26343600 DOI: 10.1016/j.jpsychires.2015.07.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/01/2015] [Accepted: 07/16/2015] [Indexed: 01/02/2023]
Abstract
BACKGROUND Cognitive deficits have been identified as an important core feature of schizophrenia. Single nucleotide polymorphisms in the transcription factor 4 (TCF4) gene have been reported to be involved in the susceptibility to schizophrenia and be significantly related to cognitive deficits of schizophrenia and controls. This study examines whether the TCF4 rs2958182 polymorphism influences cognitive functions in chronic schizophrenia and controls. METHODS The presence of the TCF4 rs2958182 was determined in 976 patients and 420 controls using a case-control design. We assessed all the patients' psychopathology using the Positive and Negative Syndrome Scale (PANSS). Cognition was assessed in 777 patients and 399 controls by using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). RESULTS There were marginally significant differences in the TCF4 rs2958182 allelic and genotypic distributions between patients and controls (χ2 = 3.48, p = 0.062 and χ2 = 0.036, p = 0.036, respectively). Cognitive test scores were significantly lower in patients than in controls on all scales (all p < 0.001) except for the visuospatial/constructional index (p > 0.05). There were significant genotype effects on delayed memory score (p = 0.013), the RBANS total score (p = 0.028) and language score (p = 0.034). Further analysis showed that the language score significantly differed according to the genotypic groups (A/A+T/A group versus T/T group) (p = 0.007) in patients but not in controls (p > 0.05), and the delayed memory score also significantly differed according to the genotypic groups (A/A+T/A group versus T/T group) (p = 0.021) in controls but not in patients (p > 0.05). CONCLUSIONS This study found that the A allele of the TCF4 rs2958182 polymorphism was the risk allele of schizophrenia, and was associated with lower cognitive performance in language in schizophrenia and delayed memory in controls. In contrast, the T allele of this polymorphism was found to be the schizophrenia risk allele in another study in Han Chinese people.
Collapse
Affiliation(s)
- Li Hui
- Institute of Wenzhou Kangning Mental Health, Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Wen-Wang Rao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, PR China
| | - Qiong Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, PR China
| | - Changgui Kou
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, PR China
| | - Jing Qin Wu
- School of Biomedical Sciences and Pharmacy, Faculty of Health, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; Biological Psychiatry Center, Beijing HuiLongGuan Hospital, Peking University, Beijing, PR China
| | - Jin Cai He
- Department of Neurology, The First Affiliated of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Min Jie Ye
- Institute of Wenzhou Kangning Mental Health, Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Jia Hong Liu
- Institute of Wenzhou Kangning Mental Health, Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Xiao Jun Xu
- Institute of Wenzhou Kangning Mental Health, Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Ke Zheng
- Institute of Wenzhou Kangning Mental Health, Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Li Na Ruan
- Institute of Wenzhou Kangning Mental Health, Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Hong Yang Liu
- Institute of Wenzhou Kangning Mental Health, Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Wei Ming Hu
- Institute of Wenzhou Kangning Mental Health, Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Tian Nan Shao
- Department of Neurology, The First Affiliated of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Michael B AngRabanes
- Department of Psychiatry and Behavioral Sciences, Harris County Psychiatric Center, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jair C Soares
- Department of Psychiatry and Behavioral Sciences, Harris County Psychiatric Center, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiang Yang Zhang
- Biological Psychiatry Center, Beijing HuiLongGuan Hospital, Peking University, Beijing, PR China; Department of Psychiatry and Behavioral Sciences, Harris County Psychiatric Center, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
84
|
Ingason A, Giegling I, Hartmann AM, Genius J, Konte B, Friedl M, Ripke S, Sullivan PF, St. Clair D, Collier DA, O'Donovan MC, Mirnics K, Rujescu D. Expression analysis in a rat psychosis model identifies novel candidate genes validated in a large case-control sample of schizophrenia. Transl Psychiatry 2015; 5:e656. [PMID: 26460480 PMCID: PMC4930128 DOI: 10.1038/tp.2015.151] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 06/16/2015] [Accepted: 07/14/2015] [Indexed: 02/06/2023] Open
Abstract
Antagonists of the N-methyl-D-aspartate (NMDA)-type glutamate receptor induce psychosis in healthy individuals and exacerbate schizophrenia symptoms in patients. In this study we have produced an animal model of NMDA receptor hypofunction by chronically treating rats with low doses of the NMDA receptor antagonist MK-801. Subsequently, we performed an expression study and identified 20 genes showing altered expression in the brain of these rats compared with untreated animals. We then explored whether the human orthologs of these genes are associated with schizophrenia in the largest schizophrenia genome-wide association study published to date, and found evidence for association for 4 out of the 20 genes: SF3B1, FOXP1, DLG2 and VGLL4. Interestingly, three of these genes, FOXP1, SF3B1 and DLG2, have previously been implicated in neurodevelopmental disorders.
Collapse
Affiliation(s)
- A Ingason
- Department of Psychiatry, University of Halle-Wittenberg, Halle, Germany
| | - I Giegling
- Department of Psychiatry, University of Halle-Wittenberg, Halle, Germany
| | - A M Hartmann
- Department of Psychiatry, University of Halle-Wittenberg, Halle, Germany
| | - J Genius
- Department of Psychiatry, Ludwig-Maximilians-University, Munich, Germany
| | - B Konte
- Department of Psychiatry, University of Halle-Wittenberg, Halle, Germany
| | - M Friedl
- Department of Psychiatry, University of Halle-Wittenberg, Halle, Germany
| | | | - S Ripke
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - P F Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - D St. Clair
- Department of Mental Health, University of Aberdeen, Aberdeen, UK
| | - D A Collier
- King's College London, Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, London, UK
| | - M C O'Donovan
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - K Mirnics
- Department of Psychiatry, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, USA
| | - D Rujescu
- Department of Psychiatry, University of Halle-Wittenberg, Halle, Germany,Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Julius-Kühn-Strasse 7, Halle 06112, Germany. E-mail:
| |
Collapse
|
85
|
Jiang H, Qiao F, Li Z, Zhang Y, Cheng Y, Xu X, Yu L. Evaluating the association between CACNA1C rs1006737 and schizophrenia risk: A meta-analysis. Asia Pac Psychiatry 2015; 7:260-7. [PMID: 25588813 DOI: 10.1111/appy.12173] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/25/2014] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Genetic analyses including genome-wide association studies have reported an intronic single nucleotide polymorphism (SNP) rs1006737 in CACNA1C gene (encoded calcium channel, voltage-dependent, L type, alpha 1C subunit) as a risk factor for schizophrenia in European populations. The replications in other ethnic populations such as East Asians have also been conducted, but the results were inconsistent, either likely due to the limited sample size of single study or genetic heterogeneity between continental populations on this locus. METHODS We performed a comprehensive meta-analysis of all available samples from existing studies of East Asian populations, including a total of 9,432 cases and 10,661 controls, to further confirm whether CACNA1C rs1006737 is an authentic risk SNP for schizophrenia in East Asian populations. RESULTS Our results revealed a significant association between rs1006737 and schizophrenia (allelic model, P = 4.39 × 10(-6) , pooled odds ratio [OR] = 1.20), and the results were much strengthened when the European and East Asian samples were combined together (P = 2.40 × 10(-17) , pooled OR = 1.12). There is no significant heterogeneity or publication bias between individual studies, and removal of any single study still remained significant associations between rs1006737 and schizophrenia. DISCUSSION Our results further confirmed that rs1006737 should be categorized as an authentic risk SNP for schizophrenia in the general populations.
Collapse
Affiliation(s)
- Hongyan Jiang
- Laboratory for Conservation and Utilization of Bio-resource, Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, Yunnan, China.,Department of Psychiatry, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fei Qiao
- Department of Anesthesiology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zongfang Li
- Department of Radiology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yaping Zhang
- Laboratory for Conservation and Utilization of Bio-resource, Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, Yunnan, China.,State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yuqi Cheng
- Department of Psychiatry, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiufeng Xu
- Department of Psychiatry, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Li Yu
- Laboratory for Conservation and Utilization of Bio-resource, Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, Yunnan, China
| |
Collapse
|
86
|
Liu X, Shimada T, Otowa T, Wu YY, Kawamura Y, Tochigi M, Iwata Y, Umekage T, Toyota T, Maekawa M, Iwayama Y, Suzuki K, Kakiuchi C, Kuwabara H, Kano Y, Nishida H, Sugiyama T, Kato N, Chen CH, Mori N, Yamada K, Yoshikawa T, Kasai K, Tokunaga K, Sasaki T, Gau SSF. Genome-wide Association Study of Autism Spectrum Disorder in the East Asian Populations. Autism Res 2015; 9:340-9. [PMID: 26314684 DOI: 10.1002/aur.1536] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/07/2015] [Accepted: 07/29/2015] [Indexed: 12/29/2022]
Abstract
Autism spectrum disorder is a heterogeneous neurodevelopmental disorder with strong genetic basis. To identify common genetic variations conferring the risk of ASD, we performed a two-stage genome-wide association study using ASD family and healthy control samples obtained from East Asian populations. A total of 166 ASD families (n = 500) and 642 healthy controls from the Japanese population were used as the discovery cohort. Approximately 900,000 single nucleotide polymorphisms (SNPs) were genotyped using Affymetrix Genome-Wide Human SNP array 6.0 chips. In the replication stage, 205 Japanese ASD cases and 184 healthy controls, as well as 418 Chinese Han trios (n = 1,254), were genotyped by TaqMan platform. Case-control analysis, family based association test, and transmission/disequilibrium test (TDT) were then conducted to test the association. In the discovery stage, significant associations were suggested for 14 loci, including 5 known ASD candidate genes: GPC6, JARID2, YTHDC2, CNTN4, and CSMD1. In addition, significant associations were identified for several novel genes with intriguing functions, such as JPH3, PTPRD, CUX1, and RIT2. After a meta-analysis combining the Japanese replication samples, the strongest signal was found at rs16976358 (P = 6.04 × 10(-7)), which is located near the RIT2 gene. In summary, our results provide independent support to known ASD candidate genes and highlight a number of novel genes warranted to be further investigated in a larger sample set in an effort to improve our understanding of the genetic basis of ASD.
Collapse
Affiliation(s)
- Xiaoxi Liu
- Department of Human Genetics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Takafumi Shimada
- Department of Neuropsychiatry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Takeshi Otowa
- Department of Neuropsychiatry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yu-Yu Wu
- Department of Psychiatry, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Yoshiya Kawamura
- Department of Psychiatry, Sakae Seijinkai Hospital, Kanagawa, Japan
| | - Mamoru Tochigi
- Department of Neuropsychiatry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yasuhide Iwata
- Department of Psychiatry and Neurology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tadashi Umekage
- Department of Neuropsychiatry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tomoko Toyota
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - Motoko Maekawa
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - Yoshimi Iwayama
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - Katsuaki Suzuki
- Department of Psychiatry and Neurology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Chihiro Kakiuchi
- Department of Neuropsychiatry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hitoshi Kuwabara
- Department of Child Psychiatry, University of Tokyo Hospital, Tokyo, Japan
| | - Yukiko Kano
- Department of Child Psychiatry, University of Tokyo Hospital, Tokyo, Japan
| | - Hisami Nishida
- Asunaro Hospital for Child and Adolescent Psychiatry, Tsu, Japan
| | - Toshiro Sugiyama
- Department of Child and Adolescent Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Nobumasa Kato
- Department of Psychiatry, Graduate School of Medicine, University of Showa, Tokyo, Japan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan.,Department and Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Norio Mori
- Department of Child and Adolescent Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuo Yamada
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tsukasa Sasaki
- Department of Physical and Health Education, Graduate School of Education, The University of Tokyo, Tokyo, Japan
| | - Susan Shur-Fen Gau
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| |
Collapse
|
87
|
Suárez-Rama JJ, Arrojo M, Sobrino B, Amigo J, Brenlla J, Agra S, Paz E, Brión M, Carracedo Á, Páramo M, Costas J. Resequencing and association analysis of coding regions at twenty candidate genes suggest a role for rare risk variation at AKAP9 and protective variation at NRXN1 in schizophrenia susceptibility. J Psychiatr Res 2015; 66-67:38-44. [PMID: 25943950 DOI: 10.1016/j.jpsychires.2015.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/10/2015] [Accepted: 04/14/2015] [Indexed: 12/20/2022]
Abstract
A fraction of genetic risk to develop schizophrenia may be due to low-frequency variants. This multistep study attempted to find low-frequency variants of high effect at coding regions of eleven schizophrenia susceptibility genes supported by genome-wide association studies (GWAS) and nine genes for the DISC1 interactome, a susceptibility gene-set. During the discovery step, a total of 125 kb per sample were resequenced in 153 schizophrenia patients and 153 controls from Galicia (NW Spain), and the cumulative role of low-frequency variants at a gene or at the DISC1 gene-set were analyzed by burden and variance-based tests. Relevant results were meta-analyzed when appropriate data were available. In addition, case-only putative damaging variants were genotyped in a further 419 cases and 398 controls. The discovery step revealed a protective effect of rare missense variants at NRXN1, a result supported by meta-analysis (OR = 0.67, 95% CI: 0.47-0.94, P = 0.021, based on 3848 patients and 3896 controls from six studies). The follow-up step based on case-only putative damaging variants revealed a promising risk variant at AKAP9. This variant, K873R, reached nominal significance after inclusion of 240 additional Spanish controls from databases. The variant, located in an ADCY2 binding region, is absent from large public databases. Interestingly, GWAS revealed an association between common ADCY2 variants and bipolar disorder, a disorder with considerable genetic overlap with schizophrenia. These data suggest a role of rare missense variants at NRXN1 and AKAP9 in schizophrenia susceptibility, probably related to alteration of the excitatory/inhibitory synaptic balance, deserving further investigation.
Collapse
Affiliation(s)
- José Javier Suárez-Rama
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Arrojo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Beatriz Sobrino
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Fundación Pública Galega de Medicina Xenómica, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Jorge Amigo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Julio Brenlla
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago(CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Santiago Agra
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago(CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Eduardo Paz
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago(CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - María Brión
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Ángel Carracedo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Fundación Pública Galega de Medicina Xenómica, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Mario Páramo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago(CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Javier Costas
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain.
| |
Collapse
|
88
|
Basmanav FB, Forstner AJ, Fier H, Herms S, Meier S, Degenhardt F, Hoffmann P, Barth S, Fricker N, Strohmaier J, Witt SH, Ludwig M, Schmael C, Moebus S, Maier W, Mössner R, Rujescu D, Rietschel M, Lange C, Nöthen MM, Cichon S. Investigation of the role of TCF4 rare sequence variants in schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2015; 168B:354-62. [PMID: 26010163 DOI: 10.1002/ajmg.b.32318] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 04/13/2015] [Indexed: 12/20/2022]
Abstract
Transcription factor 4 (TCF4) is one of the most robust of all reported schizophrenia risk loci and is supported by several genetic and functional lines of evidence. While numerous studies have implicated common genetic variation at TCF4 in schizophrenia risk, the role of rare, small-sized variants at this locus-such as single nucleotide variants and short indels which are below the resolution of chip-based arrays requires further exploration. The aim of the present study was to investigate the association between rare TCF4 sequence variants and schizophrenia. Exon-targeted resequencing was performed in 190 German schizophrenia patients. Six rare variants at the coding exons and flanking sequences of the TCF4 gene were identified, including two missense variants and one splice site variant. These six variants were then pooled with nine additional rare variants identified in 379 European participants of the 1000 Genomes Project, and all 15 variants were genotyped in an independent German sample (n = 1,808 patients; n = 2,261 controls). These data were then analyzed using six statistical methods developed for the association analysis of rare variants. No significant association (P < 0.05) was found. However, the results from our association and power analyses suggest that further research into the possible involvement of rare TCF4 sequence variants in schizophrenia risk is warranted by the assessment of larger cohorts with higher statistical power to identify rare variant associations.
Collapse
Affiliation(s)
- F Buket Basmanav
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, Bonn, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, Bonn, Germany
| | - Heide Fier
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, Bonn, Germany.,Department of Genomic Mathematics, University of Bonn, Bonn, Germany
| | - Stefan Herms
- Department of Genomics, Life and Brain Center, Bonn, Germany.,Division of Medical Genetics, University Hospital Basel and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Sandra Meier
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany.,National Center for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, Bonn, Germany
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, Bonn, Germany.,Division of Medical Genetics, University Hospital Basel and Department of Biomedicine, University of Basel, Basel, Switzerland.,Institute of Neuroscience and Medicine INM-1, Research Center Juelich, Juelich, Germany
| | - Sandra Barth
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, Bonn, Germany
| | - Nadine Fricker
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, Bonn, Germany
| | - Jana Strohmaier
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Michael Ludwig
- Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Christine Schmael
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Susanne Moebus
- Centre of Urban Epidemiology, Institute of Medical Informatics, Biometry and Epidemiology, Essen, Germany
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Rainald Mössner
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany.,Department of Psychiatry, University of Tübingen
| | - Dan Rujescu
- Department of Psychiatry, University of Halle, Halle, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Christoph Lange
- Department of Genomic Mathematics, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, Bonn, Germany
| | - Sven Cichon
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, Bonn, Germany.,Division of Medical Genetics, University Hospital Basel and Department of Biomedicine, University of Basel, Basel, Switzerland.,Institute of Neuroscience and Medicine INM-1, Research Center Juelich, Juelich, Germany
| |
Collapse
|
89
|
Association between ANKK1 (rs1800497) and LTA (rs909253) Genetic Variants and Risk of Schizophrenia. BIOMED RESEARCH INTERNATIONAL 2015. [PMID: 26114114 DOI: 10.1155/2015/821827]] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Limited research has assessed associations between schizophrenia and genetic variants of the ankyrin repeat and kinase domain containing 1 (ANKK1) and lymphotoxin-alpha (LTA) genes among individuals of Middle Eastern ancestry. Here we present the first association study investigating the ANKK1 rs1800497 (T>C) and LTA rs909253 (A>G) single-nucleotide polymorphisms in an Egyptian population. Among 120 patients with DSM-IV and PANSS (Positive and Negative Syndrome Scale) assessments of schizophrenia and 100 healthy controls, we determined the genotypes for the polymorphisms using endonuclease digestion of amplified genomic DNA. Results confirmed previous findings from different ethnic populations, in that the rs1800497 and rs909253 polymorphisms were both associated with risk of schizophrenia. Differences between the genotypes of cases and controls were strongly significant (P = 0.0005 for rs1800497 and P = 0.001 for rs909253). The relative risk to schizophrenia was 1.2 (P = 0.01) for the C allele and 0.8 (P = 0.04) for the G allele. The CC, GG, and combined CC/AA genotypes were all more frequent in cases than in controls. These results support an association between ANKK1 and LTA genetic markers and vulnerability to schizophrenia and show the potential influence of just one copy of the mutant C or G allele in the Egyptian population.
Collapse
|
90
|
Arab AH, Elhawary NA. Association between ANKK1 (rs1800497) and LTA (rs909253) Genetic Variants and Risk of Schizophrenia. BIOMED RESEARCH INTERNATIONAL 2015; 2015:821827. [PMID: 26114114 PMCID: PMC4465678 DOI: 10.1155/2015/821827] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/18/2015] [Indexed: 02/05/2023]
Abstract
Limited research has assessed associations between schizophrenia and genetic variants of the ankyrin repeat and kinase domain containing 1 (ANKK1) and lymphotoxin-alpha (LTA) genes among individuals of Middle Eastern ancestry. Here we present the first association study investigating the ANKK1 rs1800497 (T>C) and LTA rs909253 (A>G) single-nucleotide polymorphisms in an Egyptian population. Among 120 patients with DSM-IV and PANSS (Positive and Negative Syndrome Scale) assessments of schizophrenia and 100 healthy controls, we determined the genotypes for the polymorphisms using endonuclease digestion of amplified genomic DNA. Results confirmed previous findings from different ethnic populations, in that the rs1800497 and rs909253 polymorphisms were both associated with risk of schizophrenia. Differences between the genotypes of cases and controls were strongly significant (P = 0.0005 for rs1800497 and P = 0.001 for rs909253). The relative risk to schizophrenia was 1.2 (P = 0.01) for the C allele and 0.8 (P = 0.04) for the G allele. The CC, GG, and combined CC/AA genotypes were all more frequent in cases than in controls. These results support an association between ANKK1 and LTA genetic markers and vulnerability to schizophrenia and show the potential influence of just one copy of the mutant C or G allele in the Egyptian population.
Collapse
Affiliation(s)
- Arwa H. Arab
- Department of Psychology, Faculty of Arts and Humanities, King Abdulaziz University, P.O. Box 80200, Jeddah 21589, Saudi Arabia
| | - Nasser A. Elhawary
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, P.O. Box 57543, Mecca 21955, Saudi Arabia
- Department of Molecular Genetics, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| |
Collapse
|
91
|
Pierson E, Koller D, Battle A, Mostafavi S. Sharing and Specificity of Co-expression Networks across 35 Human Tissues. PLoS Comput Biol 2015; 11:e1004220. [PMID: 25970446 PMCID: PMC4430528 DOI: 10.1371/journal.pcbi.1004220] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/02/2015] [Indexed: 01/21/2023] Open
Abstract
To understand the regulation of tissue-specific gene expression, the GTEx Consortium generated RNA-seq expression data for more than thirty distinct human tissues. This data provides an opportunity for deriving shared and tissue specific gene regulatory networks on the basis of co-expression between genes. However, a small number of samples are available for a majority of the tissues, and therefore statistical inference of networks in this setting is highly underpowered. To address this problem, we infer tissue-specific gene co-expression networks for 35 tissues in the GTEx dataset using a novel algorithm, GNAT, that uses a hierarchy of tissues to share data between related tissues. We show that this transfer learning approach increases the accuracy with which networks are learned. Analysis of these networks reveals that tissue-specific transcription factors are hubs that preferentially connect to genes with tissue specific functions. Additionally, we observe that genes with tissue-specific functions lie at the peripheries of our networks. We identify numerous modules enriched for Gene Ontology functions, and show that modules conserved across tissues are especially likely to have functions common to all tissues, while modules that are upregulated in a particular tissue are often instrumental to tissue-specific function. Finally, we provide a web tool, available at mostafavilab.stat.ubc.ca/GNAT, which allows exploration of gene function and regulation in a tissue-specific manner.
Collapse
Affiliation(s)
- Emma Pierson
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| | | | - Daphne Koller
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| | - Alexis Battle
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| | - Sara Mostafavi
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| |
Collapse
|
92
|
Luo XJ, Huang L, van den Oord EJ, Aberg KA, Gan L, Zhao Z, Yao YG. Common variants in the MKL1 gene confer risk of schizophrenia. Schizophr Bull 2015; 41:715-27. [PMID: 25380769 PMCID: PMC4393692 DOI: 10.1093/schbul/sbu156] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Genome-wide association studies (GWAS) of schizophrenia have identified multiple risk variants with robust association signals for schizophrenia. However, these variants could explain only a small proportion of schizophrenia heritability. Furthermore, the effect size of these risk variants is relatively small (eg, most of them had an OR less than 1.2), suggesting that additional risk variants may be detected when increasing sample size in analysis. Here, we report the identification of a genome-wide significant schizophrenia risk locus at 22q13.1 by combining 2 large-scale schizophrenia cohort studies. Our meta-analysis revealed that 7 single nucleotide polymorphism (SNPs) on chromosome 22q13.1 reached the genome-wide significance level (P < 5.0×10(-8)) in the combined samples (a total of 38441 individuals). Among them, SNP rs6001946 had the most significant association with schizophrenia (P = 2.04×10(-8)). Interestingly, all 7 SNPs are in high linkage disequilibrium and located in the MKL1 gene. Expression analysis showed that MKL1 is highly expressed in human and mouse brains. We further investigated functional links between MKL1 and proteins encoded by other schizophrenia susceptibility genes in the whole human protein interaction network. We found that MKL1 physically interacts with GSK3B, a protein encoded by a well-characterized schizophrenia susceptibility gene. Collectively, our results revealed that genetic variants in MKL1 might confer risk to schizophrenia. Further investigation of the roles of MKL1 in the pathogenesis of schizophrenia is warranted.
Collapse
Affiliation(s)
- Xiong-jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China;,*To whom correspondence should be addressed; Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; tel: 86-871-65180085, fax: 86-871-65180085, e-mail:
| | - Liang Huang
- First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Edwin J. van den Oord
- Center for Biomarker Research and Personalized Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Karolina A. Aberg
- Center for Biomarker Research and Personalized Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Lin Gan
- Flaum Eye Institute and Department of Ophthalmology, University of Rochester, Rochester, NY 14642, USA
| | - Zhongming Zhao
- Departments of Biomedical Informatics and Psychiatry, Vanderbilt University School of Medicine, Nashville, TN
| | - Yong-Gang Yao
- 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
|
93
|
Gurung R, Prata DP. What is the impact of genome-wide supported risk variants for schizophrenia and bipolar disorder on brain structure and function? A systematic review. Psychol Med 2015; 45:2461-2480. [PMID: 25858580 DOI: 10.1017/s0033291715000537] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The powerful genome-wide association studies (GWAS) revealed common mutations that increase susceptibility for schizophrenia (SZ) and bipolar disorder (BD), but the vast majority were not known to be functional or associated with these illnesses. To help fill this gap, their impact on human brain structure and function has been examined. We systematically discuss this output to facilitate its timely integration in the psychosis research field; and encourage reflection for future research. Irrespective of imaging modality, studies addressing the effect of SZ/BD GWAS risk genes (ANK3, CACNA1C, MHC, TCF4, NRGN, DGKH, PBRM1, NCAN and ZNF804A) were included. Most GWAS risk variations were reported to affect neuroimaging phenotypes implicated in SZ/BD: white-matter integrity (ANK3 and ZNF804A), volume (CACNA1C and ZNF804A) and density (ZNF804A); grey-matter (CACNA1C, NRGN, TCF4 and ZNF804A) and ventricular (TCF4) volume; cortical folding (NCAN) and thickness (ZNF804A); regional activation during executive tasks (ANK3, CACNA1C, DGKH, NRGN and ZNF804A) and functional connectivity during executive tasks (CACNA1C and ZNF804A), facial affect recognition (CACNA1C and ZNF804A) and theory-of-mind (ZNF804A); but inconsistencies and non-replications also exist. Further efforts such as standardizing reporting and exploring complementary designs, are warranted to test the reproducibility of these early findings.
Collapse
Affiliation(s)
- R Gurung
- Department of Psychosis Studies,Institute of Psychiatry,King's College London,UK
| | - D P Prata
- Centre for Neuroimaging Sciences,Institute of Psychiatry,King's College London,UK
| |
Collapse
|
94
|
Large-scale candidate gene study to identify genetic risk factors predictive of paliperidone treatment response in patients with schizophrenia. Pharmacogenet Genomics 2015; 25:173-85. [DOI: 10.1097/fpc.0000000000000122] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
95
|
Vaccinia-Related Kinase 2 Controls the Stability of the Eukaryotic Chaperonin TRiC/CCT by Inhibiting the Deubiquitinating Enzyme USP25. Mol Cell Biol 2015; 35:1754-62. [PMID: 25755282 DOI: 10.1128/mcb.01325-14] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 02/25/2015] [Indexed: 11/20/2022] Open
Abstract
Molecular chaperones monitor the proper folding of misfolded proteins and function as the first line of defense against mutant protein aggregation in neurodegenerative diseases. The eukaryotic chaperonin TRiC is a potent suppressor of mutant protein aggregation and toxicity in early stages of disease progression. Elucidation of TRiC functional regulation will enable us to better understand the pathological mechanisms of neurodegeneration. We have previously shown that vaccinia-related kinase 2 (VRK2) downregulates TRiC protein levels through the ubiquitin-proteasome system by recruiting the E3 ligase COP1. However, although VRK2 activity was necessary in TRiC downregulation, the phosphorylated substrate was not determined. Here, we report that USP25 is a novel TRiC interacting protein that is also phosphorylated by VRK2. USP25 catalyzed deubiquitination of the TRiC protein and stabilized the chaperonin, thereby reducing accumulation of misfolded polyglutamine protein aggregates. Notably, USP25 deubiquitinating activity was suppressed when VRK2 phosphorylated the Thr(680), Thr(727), and Ser(745) residues. Impaired USP25 deubiquitinating activity after VRK2-mediated phosphorylation may be a critical pathway in TRiC protein destabilization.
Collapse
|
96
|
Cattane N, Minelli A, Milanesi E, Maj C, Bignotti S, Bortolomasi M, Chiavetto LB, Gennarelli M. Altered gene expression in schizophrenia: findings from transcriptional signatures in fibroblasts and blood. PLoS One 2015; 10:e0116686. [PMID: 25658856 PMCID: PMC4319917 DOI: 10.1371/journal.pone.0116686] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 12/12/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Whole-genome expression studies in the peripheral tissues of patients affected by schizophrenia (SCZ) can provide new insight into the molecular basis of the disorder and innovative biomarkers that may be of great utility in clinical practice. Recent evidence suggests that skin fibroblasts could represent a non-neural peripheral model useful for investigating molecular alterations in psychiatric disorders. METHODS A microarray expression study was conducted comparing skin fibroblast transcriptomic profiles from 20 SCZ patients and 20 controls. All genes strongly differentially expressed were validated by real-time quantitative PCR (RT-qPCR) in fibroblasts and analyzed in a sample of peripheral blood cell (PBC) RNA from patients (n = 25) and controls (n = 22). To evaluate the specificity for SCZ, alterations in gene expression were tested in additional samples of fibroblasts and PBCs RNA from Major Depressive Disorder (MDD) (n = 16; n = 21, respectively) and Bipolar Disorder (BD) patients (n = 15; n = 20, respectively). RESULTS Six genes (JUN, HIST2H2BE, FOSB, FOS, EGR1, TCF4) were significantly upregulated in SCZ compared to control fibroblasts. In blood, an increase in expression levels was confirmed only for EGR1, whereas JUN was downregulated; no significant differences were observed for the other genes. EGR1 upregulation was specific for SCZ compared to MDD and BD. CONCLUSIONS Our study reports the upregulation of JUN, HIST2H2BE, FOSB, FOS, EGR1 and TCF4 in the fibroblasts of SCZ patients. A significant alteration in EGR1 expression is also present in SCZ PBCs compared to controls and to MDD and BD patients, suggesting that this gene could be a specific biomarker helpful in the differential diagnosis of major psychoses.
Collapse
Affiliation(s)
- Nadia Cattane
- Department of Molecular and Translational Medicine, Biology and Genetic Division, University of Brescia, Brescia, Italy
| | - Alessandra Minelli
- Department of Molecular and Translational Medicine, Biology and Genetic Division, University of Brescia, Brescia, Italy
| | - Elena Milanesi
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Carlo Maj
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Stefano Bignotti
- Psychiatric Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Luisella Bocchio Chiavetto
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
- Faculty of Psychology, eCampus University, Novedrate, Como, Italy
| | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, Biology and Genetic Division, University of Brescia, Brescia, Italy
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
97
|
Chen J, Cao F, Liu L, Wang L, Chen X. Genetic studies of schizophrenia: an update. Neurosci Bull 2015; 31:87-98. [PMID: 25652814 DOI: 10.1007/s12264-014-1494-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/16/2014] [Indexed: 12/20/2022] Open
Abstract
Schizophrenia (SCZ) is a complex and heterogeneous mental disorder that affects about 1% of global population. In recent years, considerable progress has been made in genetic studies of SCZ. A number of common variants with small effects and rare variants with relatively larger effects have been identified. These variants include risk loci identified by genome-wide association studies, rare copy-number variants identified by comparative genomic analyses, and de novo mutations identified by high-throughput DNA sequencing. Collectively, they contribute to the heterogeneity of the disease. In this review, we update recent discoveries in the field of SCZ genetics, and outline the perspectives of future directions.
Collapse
Affiliation(s)
- Jingchun Chen
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, 23298, USA,
| | | | | | | | | |
Collapse
|
98
|
Stepanov VA, Bocharova AV, Saduakassova KZ, Marusin AV, Koneva LA, Vagaitseva KV, Svyatova GS. Replicative study of susceptibility to childhood-onset schizophrenia in Kazakhs. RUSS J GENET+ 2015; 51:185-192. [DOI: 10.1134/s1022795415020143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2025]
|
99
|
Koob AO, Shaked GM, Bender A, Bisquertt A, Rockenstein E, Masliah E. Neurogranin binds α-synuclein in the human superior temporal cortex and interaction is decreased in Parkinson's disease. Brain Res 2014; 1591:102-10. [PMID: 25446004 PMCID: PMC4943923 DOI: 10.1016/j.brainres.2014.10.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 10/06/2014] [Accepted: 10/08/2014] [Indexed: 11/26/2022]
Abstract
Neurogranin is a calmodulin binding protein that has been implicated in learning and memory, long-term potentiation and synaptic plasticity. Neurons expressing neurogranin in the cortex degenerate in late stages of Parkinson's disease with widespread α-synuclein pathology. While analyzing neurogranin gene expression levels through rtPCR in brains of mouse models overexpressing human α-synuclein, we found levels were elevated 2.5 times when compared to nontransgenic animals. Immunohistochemistry in the cortex revealed colocalization between α-synuclein and neurogranin in mouse transgenics when compared to control mice. Coimmunoprecipitation studies in the superior temporal cortex in humans confirmed interaction between α-synuclein and neurogranin, and decreased interaction between α-synuclein and neurogranin was noticed in patients diagnosed with Parkinson's disease when compared to normal control brains. Additionally, phosphorylated neurogranin levels were also decreased in the human superior temporal cortex in patients diagnosed with Parkinson's disease and patients diagnosed with dementia with Lewy bodies. Here, we show for the first time that neurogranin binds to α-synuclein in the human cortex, and this interaction decreases in Parkinson's disease along with the phosphorylation of neurogranin, a molecular process thought to be involved in learning and memory.
Collapse
Affiliation(s)
- Andrew O Koob
- Departments of Neurosciences, 9500 Gilman Drive, University of California, San Diego, La Jolla, CA 92093-0624, United States; Departments of Psychiatry, 9500 Gilman Drive, University of California, San Diego, La Jolla, CA 92093-0624, United States.
| | - Gideon M Shaked
- Departments of Neurosciences, 9500 Gilman Drive, University of California, San Diego, La Jolla, CA 92093-0624, United States
| | - Andreas Bender
- Department of Neurology, University of Munich, Klinikum der Universität München-Großhadern, 81377 München, Germany
| | - Alejandro Bisquertt
- Departments of Neurosciences, 9500 Gilman Drive, University of California, San Diego, La Jolla, CA 92093-0624, United States
| | - Edward Rockenstein
- Departments of Neurosciences, 9500 Gilman Drive, University of California, San Diego, La Jolla, CA 92093-0624, United States
| | - Eliezer Masliah
- Departments of Neurosciences, 9500 Gilman Drive, University of California, San Diego, La Jolla, CA 92093-0624, United States; Departments of Pathology, 9500 Gilman Drive, University of California, San Diego, La Jolla, CA 92093-0624, United States.
| |
Collapse
|
100
|
Mukherjee S, Guha S, Ikeda M, Iwata N, Malhotra AK, Pe'er I, Darvasi A, Lencz T. Excess of homozygosity in the major histocompatibility complex in schizophrenia. Hum Mol Genet 2014; 23:6088-95. [PMID: 24943592 PMCID: PMC4204767 DOI: 10.1093/hmg/ddu308] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 04/11/2014] [Accepted: 06/12/2014] [Indexed: 01/20/2023] Open
Abstract
Genome-wide association studies (GWAS) in schizophrenia have focused on additive allelic effects to identify disease risk loci. In order to examine potential recessive effects, we applied a novel approach to identify regions of excess homozygosity in an ethnically homogenous cohort: 904 schizophrenia cases and 1640 controls drawn from the Ashkenazi Jewish (AJ) population. Genome-wide examination of runs of homozygosity identified an excess in cases localized to the major histocompatibility complex (MHC). To refine this signal, we used the recently developed GERMLINE algorithm to identify chromosomal segments shared identical-by-descent (IBD) and compared homozygosity at such segments in cases and controls. We found a significant excess of homozygosity in schizophrenia cases compared with controls in the MHC (P-value = 0.003). An independent replication cohort of 548 schizophrenia cases from Japan and 542 matched healthy controls demonstrated similar effects. The strongest case-control recessive effects (P = 8.81 × 10(-8)) were localized to a 53-kb region near HLA-A, in a segment encompassing three poorly annotated genes, TRIM10, TRIM15 and TRIM40. At the same time, an adjacent segment in the Class I MHC demonstrated clear additive effects on schizophrenia risk, demonstrating the complexity of association in the MHC and the ability of our IBD approach to refine localization of broad signals derived from conventional GWAS. In sum, homozygosity in the classical MHC region appears to convey significant risk for schizophrenia, consistent with the ecological literature suggesting that homozygosity at the MHC locus may be associated with vulnerability to disease.
Collapse
Affiliation(s)
- Semanti Mukherjee
- The Zucker Hillside Hospital, Psychiatry Research, 75-59 263rd Street, Glen Oaks, NY 11004, USA, Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA,
| | - Saurav Guha
- The Zucker Hillside Hospital, Psychiatry Research, 75-59 263rd Street, Glen Oaks, NY 11004, USA, Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
| | - Masashi Ikeda
- Fujita Health University School of Medicine, 1-98 Kutsukake-cho Dengakugakubo, Toyoake, Aichi 470-1192, Japan
| | - Nakao Iwata
- Fujita Health University School of Medicine, 1-98 Kutsukake-cho Dengakugakubo, Toyoake, Aichi 470-1192, Japan
| | - Anil K Malhotra
- The Zucker Hillside Hospital, Psychiatry Research, 75-59 263rd Street, Glen Oaks, NY 11004, USA, Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA, Hofstra University School of Medicine, 500 Hofstra University, Hempstead, NY 11549, USA
| | - Itsik Pe'er
- Department of Computer Science, Columbia University, New York, NY 10027, USA and
| | - Ariel Darvasi
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Todd Lencz
- The Zucker Hillside Hospital, Psychiatry Research, 75-59 263rd Street, Glen Oaks, NY 11004, USA, Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA, Hofstra University School of Medicine, 500 Hofstra University, Hempstead, NY 11549, USA,
| |
Collapse
|