51
|
Cariaga-Martinez A, Saiz-Ruiz J, Alelú-Paz R. From Linkage Studies to Epigenetics: What We Know and What We Need to Know in the Neurobiology of Schizophrenia. Front Neurosci 2016; 10:202. [PMID: 27242407 PMCID: PMC4862989 DOI: 10.3389/fnins.2016.00202] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/25/2016] [Indexed: 01/15/2023] Open
Abstract
Schizophrenia is a complex psychiatric disorder characterized by the presence of positive, negative, and cognitive symptoms that lacks a unifying neuropathology. In the present paper, we will review the current understanding of molecular dysregulation in schizophrenia, including genetic and epigenetic studies. In relation to the latter, basic research suggests that normal cognition is regulated by epigenetic mechanisms and its dysfunction occurs upon epigenetic misregulation, providing new insights into missing heritability of complex psychiatric diseases, referring to the discrepancy between epidemiological heritability and the proportion of phenotypic variation explained by DNA sequence difference. In schizophrenia the absence of consistently replicated genetic effects together with evidence for lasting changes in gene expression after environmental exposures suggest a role of epigenetic mechanisms. In this review we will focus on epigenetic modifications as a key mechanism through which environmental factors interact with individual's genetic constitution to affect risk of psychotic conditions throughout life.
Collapse
Affiliation(s)
- Ariel Cariaga-Martinez
- Laboratory for Neuroscience of Mental Disorders Elena Pessino, Department of Medicine and Medical Specialties, School of Medicine, Alcalá University Madrid, Spain
| | - Jerónimo Saiz-Ruiz
- Department of Psychiatry, Ramón y Cajal Hospital, IRYCISMadrid, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM)Madrid, Spain
| | - Raúl Alelú-Paz
- Laboratory for Neuroscience of Mental Disorders Elena Pessino, Department of Medicine and Medical Specialties, School of Medicine, Alcalá UniversityMadrid, Spain; Department of Psychiatry, Ramón y Cajal Hospital, IRYCISMadrid, Spain
| |
Collapse
|
52
|
Shin C, Han C, Pae CU, Patkar AA. Precision medicine for psychopharmacology: a general introduction. Expert Rev Neurother 2016; 16:831-9. [PMID: 27104961 DOI: 10.1080/14737175.2016.1182022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Precision medicine is an emerging medical model that can provide accurate diagnoses and tailored therapeutic strategies for patients based on data pertaining to genes, microbiomes, environment, family history and lifestyle. AREAS COVERED Here, we provide basic information about precision medicine and newly introduced concepts, such as the precision medicine ecosystem and big data processing, and omics technologies including pharmacogenomics, pharamacometabolomics, pharmacoproteomics, pharmacoepigenomics, connectomics and exposomics. The authors review the current state of omics in psychiatry and the future direction of psychopharmacology as it moves towards precision medicine. Expert commentary: Advances in precision medicine have been facilitated by achievements in multiple fields, including large-scale biological databases, powerful methods for characterizing patients (such as genomics, proteomics, metabolomics, diverse cellular assays, and even social networks and mobile health technologies), and computer-based tools for analyzing large amounts of data.
Collapse
Affiliation(s)
- Cheolmin Shin
- a Department of Psychiatry, College of Medicine , Korea University , Seoul , South Korea
| | - Changsu Han
- a Department of Psychiatry, College of Medicine , Korea University , Seoul , South Korea
| | - Chi-Un Pae
- b Department of Psychiatry , Catholic University College of Medicine , Seoul , South Korea
| | - Ashwin A Patkar
- c Department of Psychiatry and Behavioural Sciences , Duke University Medical Center , Durham , NC , USA
| |
Collapse
|
53
|
The identification of novel genetic variants associated with antipsychotic treatment response outcomes in first-episode schizophrenia patients. Pharmacogenet Genomics 2016; 26:235-42. [DOI: 10.1097/fpc.0000000000000213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
54
|
Meng Q, Ying Z, Noble E, Zhao Y, Agrawal R, Mikhail A, Zhuang Y, Tyagi E, Zhang Q, Lee JH, Morselli M, Orozco L, Guo W, Kilts TM, Zhu J, Zhang B, Pellegrini M, Xiao X, Young MF, Gomez-Pinilla F, Yang X. Systems Nutrigenomics Reveals Brain Gene Networks Linking Metabolic and Brain Disorders. EBioMedicine 2016; 7:157-66. [PMID: 27322469 PMCID: PMC4909610 DOI: 10.1016/j.ebiom.2016.04.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/05/2016] [Accepted: 04/07/2016] [Indexed: 12/30/2022] Open
Abstract
Nutrition plays a significant role in the increasing prevalence of metabolic and brain disorders. Here we employ systems nutrigenomics to scrutinize the genomic bases of nutrient–host interaction underlying disease predisposition or therapeutic potential. We conducted transcriptome and epigenome sequencing of hypothalamus (metabolic control) and hippocampus (cognitive processing) from a rodent model of fructose consumption, and identified significant reprogramming of DNA methylation, transcript abundance, alternative splicing, and gene networks governing cell metabolism, cell communication, inflammation, and neuronal signaling. These signals converged with genetic causal risks of metabolic, neurological, and psychiatric disorders revealed in humans. Gene network modeling uncovered the extracellular matrix genes Bgn and Fmod as main orchestrators of the effects of fructose, as validated using two knockout mouse models. We further demonstrate that an omega-3 fatty acid, DHA, reverses the genomic and network perturbations elicited by fructose, providing molecular support for nutritional interventions to counteract diet-induced metabolic and brain disorders. Our integrative approach complementing rodent and human studies supports the applicability of nutrigenomics principles to predict disease susceptibility and to guide personalized medicine. Fructose promotes transcriptomic and epigenomic reprogramming to perturb brain networks linking metabolism and brain function. The extracellular matrix genes Bgn and Fmod emerge as key regulators of gene networks responsive to fructose. The omega-3 fatty acid DHA reverses fructose-induced genomic and network reprogramming.
Meng et al. report fructose as a powerful inducer of genomic and epigenomic variability with the capacity to reorganize gene networks critical for central metabolic regulation and neuronal processes in the brain; conversely, an omega-3 fatty acid, DHA, has the potential to normalize the genomic impact of fructose. Our findings help explain the pathogenic actions of fructose on prevalent metabolic and brain disorders and provide proof-of-concept for nutritional remedies supported by nutrigenomics evidence. Our integrative approach complementing rodent and human studies supports the applicability of nutrigenomics principles to predict disease susceptibility and to guide personalized medicine.
Collapse
Affiliation(s)
- Qingying Meng
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zhe Ying
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Emily Noble
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yuqi Zhao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rahul Agrawal
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andrew Mikhail
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yumei Zhuang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ethika Tyagi
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Qing Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jae-Hyung Lee
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Maxillofacial Biomedical Engineering, School of Dentistry, Kyung Hee University, Seoul 130-701, Korea
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Luz Orozco
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Weilong Guo
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Synthetic & Systems Biology, TNLIST, Tsinghua University, Beijing 100084, China
| | - Tina M Kilts
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York 10029, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marian F Young
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
55
|
The GRM7 gene, early response to risperidone, and schizophrenia: a genome-wide association study and a confirmatory pharmacogenetic analysis. THE PHARMACOGENOMICS JOURNAL 2016; 17:146-154. [DOI: 10.1038/tpj.2015.90] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 08/26/2015] [Accepted: 10/16/2015] [Indexed: 02/07/2023]
|
56
|
Li Z, Xiang Y, Chen J, Li Q, Shen J, Liu Y, Li W, Xing Q, Wang Q, Wang L, Feng G, He L, Zhao X, Shi Y. Loci with genome-wide associations with schizophrenia in the Han Chinese population. Br J Psychiatry 2015. [PMID: 26206863 DOI: 10.1192/bjp.bp.114.150490] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND A large schizophrenia genome-wide association study (GWAS) and a subsequent extensive replication study of individuals of European ancestry identified eight new loci with genome-wide significance and suggested that the MIR137-mediated pathway plays a role in the predisposition for schizophrenia. AIMS To validate the above findings in a Han Chinese population. METHOD We analysed the single nucleotide polymorphisms (SNPs) in the newly identified schizophrenia candidate loci and predicted MIR137 target genes based on our published Han Chinese populations (BIOX) GWAS data. We then analysed 18 SNPs from the candidate regions in an independent cohort that consisted of 3585 patients with schizophrenia and 5496 controls of Han Chinese ancestry. RESULTS We replicated the associations of five markers (P<0.05), including three that were located in the predicted MIR137 target genes. Two loci (ITIH3/4: rs2239547, P = 1.17 × 10(-10) and CALN1: rs2944829, P = 9.97 × 10(-9)) exhibited genome-wide significance in the Han Chinese population. CONCLUSIONS The ITIH3/4 locus has been reported to be of genome-wide significance in the European population. The successful replication of this finding in a different ethnic group provides stronger evidence for the association between schizophrenia and ITIH3/4. We detected the first genome-wide significant association of schizophrenia with CALN1, which is a predicted target of MIR137, and thus provide new evidence for the associations between MIR137 targets and schizophrenia.
Collapse
Affiliation(s)
- Zhiqiang Li
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yuqian Xiang
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jianhua Chen
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Qiaoli Li
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jiawei Shen
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yun Liu
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Wenjin Li
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Qinghe Xing
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Qingzhong Wang
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Lei Wang
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Guoyin Feng
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Lin He
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xinzhi Zhao
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yongyong Shi
- Zhiqiang Li, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yuqian Xiang, PhD, Children's Hospital, Fudan University, Shanghai; Jianhua Chen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qiaoli Li, PhD, Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai; Jiawei Shen, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Yun Liu, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Wenjin Li, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Qinghe Xing, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Qingzhong Wang, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Lei Wang, PhD, Institutes of Biomedical Sciences, Fudan University, Shanghai; Guoyin Feng, Shanghai Institute of Mental Health, Shanghai; Lin He, PhD, Children's Hospital, Fudan University, Shanghai and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai; Xinzhi Zhao, PhD, Children's Hospital, Fudan University, Shanghai; Yongyong Shi, PhD, Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
57
|
van Mierlo HC, van Coevorden-Hameete MH, Munting LP, de Graaff E, de Witte L. No evidence for the presence of neuronal surface autoantibodies in plasma of patients with schizophrenia. Eur Neuropsychopharmacol 2015; 25:2326-32. [PMID: 26482911 DOI: 10.1016/j.euroneuro.2015.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/05/2015] [Accepted: 09/24/2015] [Indexed: 11/18/2022]
Abstract
The immune system has been implicated in the etiology of schizophrenia. Autoimmunity by antibodies against neuronal cell surface antigens has been proposed as one of the pathological mechanisms. We examined plasma samples of 104 patients diagnosed with schizophrenia for the presence of autoantibodies against neuronal cell surface antigens using cultured hippocampal neurons and transfected HeLa cells. None of the samples tested positive for the presence of these autoantibodies. Based on our results it seems unlikely that autoantibodies against neuronal cell surface antigens play a role in the pathogenesis of schizophrenia, although further studies using cerebrospinal fluid are needed.
Collapse
Affiliation(s)
- Hans C van Mierlo
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | - Leon P Munting
- Department of Biology, Division of Cell Biology, Utrecht University, The Netherlands
| | - Esther de Graaff
- Department of Biology, Division of Cell Biology, Utrecht University, The Netherlands
| | - Lot de Witte
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
58
|
New discoveries in schizophrenia genetics reveal neurobiological pathways: A review of recent findings. Eur J Med Genet 2015; 58:704-14. [PMID: 26493318 DOI: 10.1016/j.ejmg.2015.10.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/14/2015] [Accepted: 10/15/2015] [Indexed: 02/08/2023]
Abstract
Schizophrenia research has undergone a recent transformation. By leveraging large sample sizes, genome-wide association studies of common genetic variants have approximately tripled the number of candidate genetic loci. Rare variant studies have identified copy number variants that are schizophrenia risk loci. Among these, the 3q29 microdeletion is now known to be the single largest schizophrenia risk factor. Next-generation sequencing studies are increasingly used for rare variant association testing, and have already facilitated identification of large effect alleles. Collectively, recent findings implicate voltage-gated calcium channel and cytoskeletal pathways in the pathogenesis of schizophrenia. Taken together, these results suggest the possibility of imminent breakthroughs in the molecular understanding of schizophrenia.
Collapse
|
59
|
Meta-analysis of data from the Psychiatric Genomics Consortium and additional samples supports association of CACNA1C with risk for schizophrenia. Schizophr Res 2015; 168:429-33. [PMID: 26276307 DOI: 10.1016/j.schres.2015.07.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 07/10/2015] [Accepted: 07/18/2015] [Indexed: 12/18/2022]
Abstract
Recently, numerous genome-wide association studies (GWASs) have identified numerous risk loci for schizophrenia, but follow-up studies are still essential to confirm those results. Therefore, we followed up on top GWAS hits by genotyping implicated loci in additional schizophrenia family samples from our own collection. Five-hundred thirty-six Asian families (comprising 1633 members including 698 schizophrenics) were genotyped in this study. We analyzed 12 single nucleotide polymorphisms (SNPs) in strongly implicated candidate genes revealed by GWASs and their follow-up studies. We then used meta-analysis to combine our results with those of the Schizophrenia Working Group of the Psychiatric Genomics Consortium (PGC). In our newly genotyped samples, there were no significant associations of any of the 12 candidate SNPs with schizophrenia; however, all genome-wide significant results from the schizophrenia PGC analysis were maintained after combination with our new data by meta-analysis. One SNP (rs4765905 in CACNA1C) showed a stronger effect and decreased p-value (5.14e-17) after meta-analysis relative to the original PGC results, with no significant between-study heterogeneity. The findings of this study support the significant results in the PGC, especially for CACNA1C. The sample size in our study was considerably smaller than that in the PGC-SCZ study; thus, the weights carried by our samples in the meta-analysis were small. Therefore, our data could not vastly reduce PGC association signals. However, we considered that the well replicated results from the PGC hold up in our new samples, and may suggest that the top hits from the PGC are generalizable, even to other ancestral groups.
Collapse
|
60
|
Dong E, Ruzicka WB, Grayson D, Guidotti A. DNA-methyltransferase1 (DNMT1) binding to CpG rich GABAergic and BDNF promoters is increased in the brain of schizophrenia and bipolar disorder patients. Schizophr Res 2015; 167:35-41. [PMID: 25476119 PMCID: PMC4451449 DOI: 10.1016/j.schres.2014.10.030] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/16/2014] [Accepted: 10/19/2014] [Indexed: 10/24/2022]
Abstract
The down regulation of glutamic acid decarboxylase67 (GAD1), reelin (RELN), and BDNF expression in brain of schizophrenia (SZ) and bipolar (BP) disorder patients is associated with overexpression of DNA methyltransferase1 (DNMT1) and ten-eleven translocase methylcytosine dioxygenase1 (TET1). DNMT1 and TET1 belong to families of enzymes that methylate and hydroxymethylate cytosines located proximal to and within cytosine phosphodiester guanine (CpG) islands of many gene promoters, respectively. Altered promoter methylation may be one mechanism underlying the down-regulation of GABAergic and glutamatergic gene expression. However, recent reports suggest that both DNMT1 and TET1 directly bind to unmethylated CpG rich promoters through their respective Zinc Finger (ZF-CXXC) domains. We report here, that the binding of DNMT1 to GABAergic (GAD1, RELN) and glutamatergic (BDNF-IX) promoters is increased in SZ and BP disorder patients and this increase does not necessarily correlate with enrichment in promoter methylation. The increased DNMT1 binding to these promoter regions is detected in the cortex but not in the cerebellum of SZ and BP disorder patients, suggesting a brain region and neuron specific dependent mechanism. Increased binding of DNMT1 positively correlates with increased expression of DNMT1 and with increased binding of MBD2. In contrast, the binding of TET1 to RELN, GAD1 and BDNF-IX promoters failed to change. These data are consistent with the hypothesis that the down-regulation of specific GABAergic and glutamatergic genes in SZ and BP disorder patients may be mediated, at least in part, by a brain region specific and neuronal-activity dependent DNMT1 action that is likely independent of its DNA methylation activity.
Collapse
Affiliation(s)
- E. Dong
- Department of Psychiatry, College of Medicine, University of Illinois at Chicago
| | - W. B. Ruzicka
- Program in Structural and Molecular Neuroscience, McLean Hospital., Belmont, MA,Department of Psychiatry, Harvard Medical School, Boston, MA
| | - D.R. Grayson
- Department of Psychiatry, College of Medicine, University of Illinois at Chicago
| | - A. Guidotti
- Department of Psychiatry, College of Medicine, University of Illinois at Chicago,Corresponding author: A. Guidotti, 1601 W. Taylor St., Chicago, IL 60612, , 312-413-4594
| |
Collapse
|
61
|
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: 80] [Impact Index Per Article: 8.0] [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
|
62
|
Bergeron SA, Carrier N, Li GH, Ahn S, Burgess HA. Gsx1 expression defines neurons required for prepulse inhibition. Mol Psychiatry 2015; 20:974-85. [PMID: 25224259 PMCID: PMC4362800 DOI: 10.1038/mp.2014.106] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 07/09/2014] [Accepted: 08/04/2014] [Indexed: 02/07/2023]
Abstract
In schizophrenia, cognitive overload is thought to reflect an inability to suppress non-salient information, a process which is studied using prepulse inhibition (PPI) of the startle response. PPI is reduced in schizophrenia and routinely tested in animal models and preclinical trials of antipsychotic drugs. However, the underlying neuronal circuitry is not well understood. We used a novel genetic screen in larval zebrafish to reveal the molecular identity of neurons that are required for PPI in fish and mice. Ablation or optogenetic silencing of neurons with developmental expression of the transcription factor genomic screen homeobox 1 (gsx1) produced profound defects in PPI in zebrafish, and PPI was similarly impaired in Gsx1 knockout mice. Gsx1-expressing neurons reside in the dorsal brainstem and form synapses closely apposed to neurons that initiate the startle response. Surprisingly, brainstem Gsx1 neurons are primarily glutamatergic despite their role in a functionally inhibitory pathway. As Gsx1 has an important role in regulating interneuron development in the forebrain, these findings reveal a molecular link between control of interneuron specification and circuits that gate sensory information across brain regions.
Collapse
Affiliation(s)
- Sadie A. Bergeron
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Nicole Carrier
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Grace H. Li
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Sohyun Ahn
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Harold A. Burgess
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA,6 Center Drive, Building 6B, Rm 3B308, Bethesda, MD 20892, , tel: 301-402-6018; fax: 301-496-0243
| |
Collapse
|
63
|
Zeledón M, Eckart N, Taub M, Vernon H, Szymanski M, Wang R, Chen PL, Nestadt G, McGrath JA, Sawa A, Pulver AE, Avramopoulos D, Valle D. Identification and functional studies of regulatory variants responsible for the association of NRG3 with a delusion phenotype in schizophrenia. MOLECULAR NEUROPSYCHIATRY 2015; 1:36-46. [PMID: 26528484 PMCID: PMC4627703 DOI: 10.1159/000371518] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/12/2014] [Indexed: 12/12/2022]
Abstract
We previously reported genetic linkage for Schizophrenia (SZ) (NPL of 4.7) at 10q22 in the Ashkenazi Jewish (AJ) population. In follow up fine mapping we found strong evidence of association between three intronic single nucleotide variants (SNVs) in the 5' end of Neuregulin 3 (NRG3) and the delusion factor score of our phenotypic principal component analysis. Two independent groups replicated these findings, indicating that variants in NRG3 confer risk for a delusion-rich SZ subtype. To identify the causative variants, we sequenced the 162 kb linkage disequilibrium (LD) block covering the NRG3 5' end in 47 AJ SZ patients at the extremes of the delusion factor quantitative trait distribution. Among the identified variants we found 5 noncoding SNVs present on the high delusion factor haplotype and significantly overrepresented in high delusion factor subjects. We tested these for regulatory effects and found that risk alleles of rs10883866 and rs60827755 decreased and increased, respectively, the expression of a reporter gene as compared to the reference allele. In post-mortem brain RNA quantification experiments we found the same variants also perturb relative expression of alternative NRG3 isoforms. In summary, we have identified regulatory SNVs contributing to the association of NRG3 with delusion symptoms in SZ.
Collapse
Affiliation(s)
- Mariela Zeledón
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Md., USA
- Predoctoral Training Program in Human Genetics, Johns Hopkins University School of Medicine, Baltimore, Md., USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Nicole Eckart
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Md., USA
- Predoctoral Training Program in Human Genetics, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Margaret Taub
- Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Md., USA
| | - Hilary Vernon
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Megan Szymanski
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Md., USA
- Predoctoral Training Program in Human Genetics, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Ruihua Wang
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Pei-Lung Chen
- Department of Medical Genetics, National Taiwan University Hospital, Taipei City, Taiwan, ROC
| | - Gerry Nestadt
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - John A. McGrath
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Md., USA
- Epidemiology-Genetics Program, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Md., USA
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Ann E. Pulver
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA
- Epidemiology-Genetics Program, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Md., USA
| | - Dimitrios Avramopoulos
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Md., USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - David Valle
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| |
Collapse
|
64
|
Lopes R, Soares R, Coelho R, Figueiredo-Braga M. Angiogenesis in the pathophysiology of schizophrenia — A comprehensive review and a conceptual hypothesis. Life Sci 2015; 128:79-93. [DOI: 10.1016/j.lfs.2015.02.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 01/27/2015] [Accepted: 02/12/2015] [Indexed: 01/11/2023]
|
65
|
Vijayakumari AA, John JP, Halahalli HN, Paul P, Thirunavukkarasu P, Purushottam M, Jain S. Effect of polymorphisms of three genes mediating monoamine signalling on brain morphometry in schizophrenia and healthy subjects. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2015; 13:68-82. [PMID: 25912540 PMCID: PMC4423152 DOI: 10.9758/cpn.2015.13.1.68] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 08/18/2014] [Accepted: 10/19/2014] [Indexed: 01/11/2023]
Abstract
OBJECTIVE We examined the effect of risk alleles of polymorphisms of three schizophrenia risk genes that mediate monoamine signalling in the brain on regional brain volumes of schizophrenia and healthy control subjects. The risk alleles and the gene polymorphisms studied were: Val allele of catechol o-methyltransferase (COMT) rs4680 polymorphism; short allele of 5-hydroxy tryptamine transporter linked polymorphic region (5HTTLPR) polymorphism; and T allele of 5-hydroxy tryptamine 2A (5HT2A) rs6314 polymorphism. METHODS The study was carried out on patients with recent onset schizophrenia (n=41) recruited from the outpatient department of National Institute of Mental Health and Neurosciences, Bangalore, India and healthy control subjects (n=39), belonging to South Indian Dravidian ethnicity. Individual and additive effects of risk alleles of the above gene polymorphisms on brain morphometry were explored using voxel-based morphometry. RESULTS Irrespective of phenotypes, individuals with the risk allele T of the rs6314 polymorphism of 5HT2A gene showed greater (at cluster-extent equivalent to family wise error-correction [FWEc] p<0.05) regional brain volumes in the left inferior temporal and left inferior occipital gyri. Those with the risk alleles of the other two polymorphisms showed a trend (at p<0.001, uncorrected) towards lower regional brain volumes. A trend (at p<0.001, uncorrected) towards additive effects of the above 3 risk alleles (subjects with 2 or 3 risk alleles vs. those with 1 or no risk alleles) on brain morphology was also noted. CONCLUSIONS The findings of the present study have implications in understanding the role of individual and additive effects of genetic variants in mediating regional brain morphometry in health and disease.
Collapse
Affiliation(s)
- Anupa A Vijayakumari
- Multimodal Brain Image Analysis Laboratory (MBIAL), India.,Departments of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - John P John
- Multimodal Brain Image Analysis Laboratory (MBIAL), India.,Departments of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India.,Departments of Clinical Neuroscience, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Harsha N Halahalli
- Departments of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Pradip Paul
- Departments of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Priyadarshini Thirunavukkarasu
- Multimodal Brain Image Analysis Laboratory (MBIAL), India.,Departments of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Meera Purushottam
- Departments of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Sanjeev Jain
- Departments of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| |
Collapse
|
66
|
Li W, Ji W, Li Z, He K, Wang Q, Chen J, Qiang Y, Feng G, Li X, Shen J, Wen Z, Ji J, Shi Y. Genetic association of ACSM1 variation with schizophrenia and major depressive disorder in the Han Chinese population. Am J Med Genet B Neuropsychiatr Genet 2015; 168B:144-9. [PMID: 25656805 DOI: 10.1002/ajmg.b.32291] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/25/2014] [Indexed: 11/08/2022]
Abstract
Schizophrenia (SCZ) and major depressive disorder (MDD) are two of the most common and severe mental disorders, the etiologies of which are not yet clearly elucidated. The ACSM1 gene has been identified as a susceptibility gene for SCZ in two previous genome-wide association studies (GWAS). ACSM1 catalyzes the activation of fatty acids and plays an important role in the metabolic system. Some evidence has suggested that ACSM1 contributes to a genetic risk for MDD. The present study aimed to evaluate the common genetic risk of the ACSM1 gene in these two disorders in the Han Chinese population. In total, 1235 patients with SCZ, 1045 patients with MDD and 1235 control subjects of Chinese origin were recruited. Six single nuclear polymorphisms (SNPs) in ACSM1 were genotyped to test their associations with SCZ and MDD. SNP rs163234 was found to be significantly associated with both SCZ (permutated Pallele=1.700×10(-3), OR=1.350 [95% CI=1.152-1.581]) and MDD (permutated Pallele=4.800×10(-3), OR=1.329 [95% CI=1.127-1.567]). SNP rs433598 showed a strong association with SCZ (permutated Pallele=4.300×10(-3), OR=1.303 [95% CI=1.117-1.520]). Haplotype analysis of the blocks containing the two positive markers also revealed a significant association. This is the first study to assess the possible association of the ACSM1 gene with a genetic susceptibility for MDD. Our data are the first to suggest a positive association of the ACSM1 gene with a genetic susceptibility for SCZ and MDD in the Han Chinese population.
Collapse
Affiliation(s)
- Wenjin Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, P.R. China; Bio-X Institutes, Key Laboratory of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, P.R. China; Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Ramsey TL, Liu Q, Brennan MD. Replication of SULT4A1-1 as a pharmacogenetic marker of olanzapine response and evidence of lower weight gain in the high response group. Pharmacogenomics 2015; 15:933-9. [PMID: 24956247 DOI: 10.2217/pgs.14.54] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Antipsychotic efficacy biomarkers have the potential to improve outcomes in psychotic patients. This study examined the effect of SULT4A1-1 haplotype status (rs2285162 [A]-rs2285167 [G]) on olanzapine response. PATIENTS & METHODS We evaluated 87 olanzapine treated subjects from Phases 1, 1B and 2 of the CATIE trial for the impact of SULT4A1-1 status on change in Positive and Negative Syndrome Scale (PANSS) total score using two models of response. We also examined weight change. RESULTS SULT4A1-1-positive status correlated with superior olanzapine response in Phase 1 (p = 0.004 for model 1 and p = 0.001 for model 2) and Phases 1B/2 (p = 0.05 for model 1 and p = 0.007 for model 2). SULT4A1-1-positive subjects gained significantly less weight per month on olanzapine, 0.15 lbs, than did SULT4A1-1-negative subjects, 2.27 lbs (p = 0.04). CONCLUSION This study provides a second replication of superior olanzapine response in SULT4A1-1-positive subjects compared with SULT4A1-1-negative subjects. SULT4A1-1-positive subjects treated with olanzapine also gained less weight than SULT4A1-1-negative subjects.
Collapse
|
68
|
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]
|
69
|
Abstract
Schizophrenia is a common mental disorder, affecting 0.5-1% of the population. The mode of inheritance is complex and non-Mendelian with a high heritability of ca. 65-80%. Given this complexity, until most recently it was difficult to identify disease genes. But fortunately this has changed. Due to new technologies the last few years have brought highest interest in human genetics of complex diseases. The knowledge resulting from the availability of the complete sequence of the human genome, the systematic identification of single nucleotide polymorphisms (SNPs) throughout the genome, and the development of parallel genotyping technology (microarrays) established the conditions that brought about the current successful time in our ability to probe the genome for identifying disease genes. All these studies showed up new avenues for the biology of common complex diseases and yielded a multitude of genes showing strong association with complex diseases.
Collapse
|
70
|
Abstract
This review considers pharmacogenetics of the so called 'second-generation' antipsychotics. Findings for polymorphisms replicating in more than one study are emphasized and compared and contrasted with larger-scale candidate gene studies and genome-wide association study analyses. Variants in three types of genes are discussed: pharmacokinetic genes associated with drug metabolism and disposition, pharmacodynamic genes encoding drug targets, and pharmacotypic genes impacting disease presentation and subtype. Among pharmacokinetic markers, CYP2D6 metabolizer phenotype has clear clinical significance, as it impacts dosing considerations for aripiprazole, iloperidone and risperidone, and variants of the ABCB1 gene hold promise as biomarkers for dosing for olanzapine and clozapine. Among pharmacodynamic variants, the TaqIA1 allele of the DRD2 gene, the DRD3 (Ser9Gly) polymorphism, and the HTR2C -759C/T polymorphism have emerged as potential biomarkers for response and/or side effects. However, large-scale candidate gene studies and genome-wide association studies indicate that pharmacotypic genes may ultimately prove to be the richest source of biomarkers for response and side effect profiles for second-generation antipsychotics.
Collapse
Affiliation(s)
- Mark D Brennan
- Department of Biochemistry & Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
71
|
Dick DM, Agrawal A, Keller MC, Adkins A, Aliev F, Monroe S, Hewitt JK, Kendler KS, Sher KJ. Candidate gene-environment interaction research: reflections and recommendations. PERSPECTIVES ON PSYCHOLOGICAL SCIENCE 2015; 10:37-59. [PMID: 25620996 PMCID: PMC4302784 DOI: 10.1177/1745691614556682] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Studying how genetic predispositions come together with environmental factors to contribute to complex behavioral outcomes has great potential for advancing the understanding of the development of psychopathology. It represents a clear theoretical advance over studying these factors in isolation. However, research at the intersection of multiple fields creates many challenges. We review several reasons why the rapidly expanding candidate gene-environment interaction (cG×E) literature should be considered with a degree of caution. We discuss lessons learned about candidate gene main effects from the evolving genetics literature and how these inform the study of cG×E. We review the importance of the measurement of the gene and environment of interest in cG×E studies. We discuss statistical concerns with modeling cG×E that are frequently overlooked. Furthermore, we review other challenges that have likely contributed to the cG×E literature being difficult to interpret, including low power and publication bias. Many of these issues are similar to other concerns about research integrity (e.g., high false-positive rates) that have received increasing attention in the social sciences. We provide recommendations for rigorous research practices for cG×E studies that we believe will advance its potential to contribute more robustly to the understanding of complex behavioral phenotypes.
Collapse
Affiliation(s)
| | - Arpana Agrawal
- Department of Psychiatry, Washington University in St. Louis
| | - Matthew C Keller
- Institute for Behavioral Genetics, University of Colorado Boulder
| | - Amy Adkins
- Department of Psychiatry, Virginia Commonwealth University
| | - Fazil Aliev
- Department of Psychiatry, Virginia Commonwealth University
| | - Scott Monroe
- Department of Psychology, University of Notre Dame
| | - John K Hewitt
- Department of Psychiatry, Washington University in St. Louis
| | | | - Kenneth J Sher
- Department of Psychological Sciences, University of Missouri
| |
Collapse
|
72
|
Affiliation(s)
- David Roofeh
- a Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Divya Tumuluru
- b Department of Psychiatry, University of Pittsburgh School of Medicine
| | - Sona Shilpakar
- b Department of Psychiatry, University of Pittsburgh School of Medicine
| | | |
Collapse
|
73
|
Stepanov VA, Bocharova AV, Marusin AV, Zhukova NG, Alifirova VM, Zhukova IA. Replicative association analysis of genetic markers of cognitive traits with Alzheimer’s disease in the Russian population. Mol Biol 2014. [DOI: 10.1134/s0026893314060168] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
74
|
Light G, Greenwood TA, Swerdlow NR, Calkins ME, Freedman R, Green MF, Gur RE, Gur RC, Lazzeroni LC, Nuechterlein KH, Olincy A, Radant AD, Seidman LJ, Siever LJ, Silverman JM, Sprock J, Stone WS, Sugar CA, Tsuang DW, Tsuang MT, Turetsky BI, Braff DL. Comparison of the heritability of schizophrenia and endophenotypes in the COGS-1 family study. Schizophr Bull 2014; 40:1404-11. [PMID: 24903414 PMCID: PMC4193725 DOI: 10.1093/schbul/sbu064] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Twin and multiplex family studies have established significant heritability for schizophrenia (SZ), often summarized as 81%. The Consortium on the Genetics of Schizophrenia (COGS-1) family study was designed to deconstruct the genetic architecture of SZ using neurocognitive and neurophysiological endophenotypes, for which heritability estimates ranged from 18% to 50% (mean = 30%). This study assessed the heritability of SZ in these families to determine whether there is a "heritability gap" between the diagnosis and related endophenotypes. METHODS Nuclear families (N = 296) with a SZ proband, an unaffected sibling, and both parents (n = 1366 subjects; mean family size = 4.6) underwent comprehensive endophenotype and clinical characterization. The Family Interview for Genetic Studies was administered to all participants and used to obtain convergent psychiatric symptom information for additional first-degree relatives of interviewed subjects (N = 3304 subjects; mean family size = 11.2). Heritability estimates of psychotic disorders were computed for both nuclear and extended families. RESULTS The heritability of SZ was 31% and 44% for nuclear and extended families. The inclusion of bipolar disorder increased the heritability to 37% for the nuclear families. When major depression was added, heritability estimates dropped to 34% and 20% for nuclear and extended families, respectively. CONCLUSIONS Endophenotypes and psychotic disorders exhibit comparable levels of heritability in the COGS-1 family sample. The ascertainment of families with discordant sibpairs to increase endophenotypic contrast may underestimate diagnostic heritability relative to other studies. However, population-based studies also report significantly lower heritability estimates for SZ. Collectively, these findings support the importance of endophenotype-based strategies and the dimensional view of psychosis.
Collapse
Affiliation(s)
- Gregory Light
- Department of Psychiatry, University of California San Diego, La Jolla, CA; VISN-22 Mental Illness, Research, Education and Clinical Center, San Diego Healthcare System La Jolla, CA;
| | - Tiffany A. Greenwood
- Department of Psychiatry, University of California San Diego, La Jolla, CA;,These authors contributed equally to the article
| | - Neal R. Swerdlow
- Department of Psychiatry, University of California San Diego, La Jolla, CA
| | - Monica E. Calkins
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Robert Freedman
- Department of Psychiatry, University of Colorado Health Sciences Center, Denver, CO
| | - Michael F. Green
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA;,VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Laura C. Lazzeroni
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA
| | - Keith H. Nuechterlein
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA
| | - Ann Olincy
- Department of Psychiatry, University of Colorado Health Sciences Center, Denver, CO
| | - Allen D. Radant
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA;,VA Puget Sound Health Care System, Seattle, WA
| | - Larry J. Seidman
- Department of Psychiatry, Harvard Medical School, Boston, MA;,Massachusetts Mental Health Center Public Psychiatry, Division of the Beth Israel Deaconess Medical Center, Boston, MA
| | - Larry J. Siever
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY;,James J. Peters VA Medical Center, New York, NY
| | - Jeremy M. Silverman
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY;,James J. Peters VA Medical Center, New York, NY
| | - Joyce Sprock
- Department of Psychiatry, University of California San Diego, La Jolla, CA
| | - William S. Stone
- Department of Psychiatry, Harvard Medical School, Boston, MA;,Massachusetts Mental Health Center Public Psychiatry, Division of the Beth Israel Deaconess Medical Center, Boston, MA
| | - Catherine A. Sugar
- Department of Biostatistics, University of California, Los Angeles School of Public Health, Los Angeles, CA
| | - Debby W. Tsuang
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA;,VA Puget Sound Health Care System, Seattle, WA
| | - Ming T. Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, CA;,Center for Behavioral Genomics, Institute for Genomic Medicine, University of California San Diego, La Jolla, CA;,Harvard Institute of Psychiatric Epidemiology and Genetics, Boston, MA
| | - Bruce I. Turetsky
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - David L. Braff
- Department of Psychiatry, University of California San Diego, La Jolla, CA
| |
Collapse
|
75
|
Zhang B, Xu YH, Wei SG, Zhang HB, Fu DK, Feng ZF, Guan FL, Zhu YS, Li SB. Association study identifying a new susceptibility gene (AUTS2) for schizophrenia. Int J Mol Sci 2014; 15:19406-16. [PMID: 25347278 PMCID: PMC4264119 DOI: 10.3390/ijms151119406] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 09/23/2014] [Accepted: 10/10/2014] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SCZ) is a severe and debilitating mental disorder, and the specific genetic factors that underlie the risk for SCZ remain elusive. The autism susceptibility candidate 2 (AUTS2) gene has been reported to be associated with autism, suicide, alcohol consumption, and heroin dependence. We hypothesized that AUTS2 might be associated with SCZ. In the present study, three polymorphisms (rs6943555, rs7459368, and rs9886351) in the AUTS2 gene were genotyped in 410 patients with SCZ and 435 controls using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) and forced PCR-RFLP methods. We detected an association between SCZ and the rs6943555 genotype distribution (odds ratio (OR)=1.363, 95% confidence interval (CI): 0.848-2.191, p=0.001). The association remained significant after adjusting for gender, and a significant effect (p=0.001) was observed among the females. In the present study, rs6943555 was determined to be associated with female SCZ. Our results confirm previous reports which have suggested that rs6943555 might elucidate the pathogenesis of schizophrenia and play an important role in its etiology.
Collapse
Affiliation(s)
- Bao Zhang
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Yue-Hong Xu
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Shu-Guang Wei
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Hong-Bo Zhang
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Dong-Ke Fu
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Zu-Fei Feng
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Fang-Lin Guan
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Yong-Sheng Zhu
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Sheng-Bin Li
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
76
|
Cunningham C, Peters K. Aetiology of schizophrenia and implications for nursing practice: a literature review. Issues Ment Health Nurs 2014; 35:732-8. [PMID: 25259635 DOI: 10.3109/01612840.2014.908441] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Schizophrenia is recognised as a major mental illness and is prevalent in approximately 1% of the global population. This paper reviewed literature published between 2001 and 2013 to explore the aetiology of schizophrenia and discuss implications for mental health nursing practice. Although the causes of schizophrenia are largely unknown, there is a strong correlation between identified physiological and environmental markers and the development of the disease. With this knowledge, mental health nurses can potentially contribute to the development of strategies and approaches to complement current treatments, and therefore ensure improved outcomes for mental health clients.
Collapse
|
77
|
Russell TA, Blizinsky KD, Cobia DJ, Cahill ME, Xie Z, Sweet RA, Duan J, Gejman PV, Wang L, Csernansky JG, Penzes P. A sequence variant in human KALRN impairs protein function and coincides with reduced cortical thickness. Nat Commun 2014; 5:4858. [PMID: 25224588 PMCID: PMC4166532 DOI: 10.1038/ncomms5858] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 07/31/2014] [Indexed: 12/16/2022] Open
Abstract
Dendritic spine pathology is a key feature of several neuropsychiatric disorders. The Rac1 guanine nucleotide exchange factor kalirin-7 is critical for spine morphogenesis on cortical pyramidal neurons. Here we identify a rare coding variant in the KALRN gene region that encodes the catalytic domain, in a schizophrenia patient and his sibling with major depressive disorder. The D1338N substitution significantly diminished the protein's ability to catalyse the activation of Rac1. Contrary to wild-type kalirin-7, kalirin-7-D1338N failed to increase spine size and density. Both subjects carrying the polymorphism displayed reduced cortical volume in the superior temporal sulcus (STS), a region implicated in schizophrenia. Consistent with this, mice with reduced kalirin expression showed reduced neuropil volume in the rodent homologue of the STS. These data suggest that single amino acid changes in proteins involved in dendritic spine function can have significant effects on the structure and function of the cerebral cortex.
Collapse
Affiliation(s)
- Theron A Russell
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Katherine D Blizinsky
- 1] Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA [2] Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Derin J Cobia
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Michael E Cahill
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Zhong Xie
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Robert A Sweet
- 1] Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA [2] Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Jubao Duan
- 1] Department of Psychiatry and Behavioral Sciences, University of Chicago, Chicago, Illinois 60637, USA [2] Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston, Illinois 60208, USA
| | - Pablo V Gejman
- 1] Department of Psychiatry and Behavioral Sciences, University of Chicago, Chicago, Illinois 60637, USA [2] Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston, Illinois 60208, USA
| | - Lei Wang
- 1] Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA [2] Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - John G Csernansky
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Peter Penzes
- 1] Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA [2] Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| |
Collapse
|
78
|
Mei L, Nave KA. Neuregulin-ERBB signaling in the nervous system and neuropsychiatric diseases. Neuron 2014; 83:27-49. [PMID: 24991953 DOI: 10.1016/j.neuron.2014.06.007] [Citation(s) in RCA: 436] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neuregulins (NRGs) comprise a large family of growth factors that stimulate ERBB receptor tyrosine kinases. NRGs and their receptors, ERBBs, have been identified as susceptibility genes for diseases such as schizophrenia (SZ) and bipolar disorder. Recent studies have revealed complex Nrg/Erbb signaling networks that regulate the assembly of neural circuitry, myelination, neurotransmission, and synaptic plasticity. Evidence indicates there is an optimal level of NRG/ERBB signaling in the brain and deviation from it impairs brain functions. NRGs/ERBBs and downstream signaling pathways may provide therapeutic targets for specific neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Lin Mei
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Goettingen, Germany.
| |
Collapse
|
79
|
Jenkins A, Apud JA, Zhang F, Decot H, Weinberger DR, Law AJ. Identification of candidate single-nucleotide polymorphisms in NRXN1 related to antipsychotic treatment response in patients with schizophrenia. Neuropsychopharmacology 2014; 39:2170-8. [PMID: 24633560 PMCID: PMC4104334 DOI: 10.1038/npp.2014.65] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 02/20/2014] [Accepted: 03/09/2014] [Indexed: 12/22/2022]
Abstract
Neurexins are presynaptic neuronal adhesion molecules that interact with postsynaptic neuroligins to form an inter-synaptic complex required for synaptic specification and efficient neurotransmission. Deletions and point mutations in the neurexin 1 (NRXN1) gene are associated with a broad spectrum of neuropsychiatric and neurodevelopmental disorders, including autism, intellectual disability, epilepsy, developmental delay, and schizophrenia. Recently, small nucleotide polymorphisms in NRXN1 have been associated with antipsychotic drug response in patients with schizophrenia. Based on previous suggestive evidence of an impact on clozapine response in patients with schizophrenia, we conducted an association study of NRXN1 polymorphisms (rs12467557 and rs10490162) with antipsychotic treatment response in 54 patients with schizophrenia in a double blind, placebo-controlled NIMH inpatient crossover trial and examined for association with risk for schizophrenia in independent case-control and family-based clinical cohorts. Pharmacogenetic analysis in the placebo controlled trial revealed significant association of rs12467557and rs10490162 with drug response, whereby individuals homozygous for the A allele, at either SNP, showed significant improvement in positive symptoms, general psychopathology, thought disturbance, and negative symptoms, whereas patients carrying the G allele showed no overall response. Although we did not find evidence of the same NRXN1 SNPs being associated with results of the NIMH sponsored CATIE trial, other SNPs showed weakly positive signals. The family and case-control analyses for schizophrenia risk were negative. Our results provide confirmatory evidence of genetically determined differences in drug response in patients with schizophrenia related to NRXN1 variation. Furthermore, these findings potentially implicate NRXN1 in the therapeutic actions of antipsychotic drugs.
Collapse
Affiliation(s)
- Aaron Jenkins
- Clinical Brain Disorders Branch, Genes, Cognition and Psychosis Program, National Institute of Mental Health, National Institute of Health, National Institutes of Health, Bethesda, MD, USA,University of Kentucky College of Medicine, Lexington, KY, USA
| | - José A Apud
- Clinical Brain Disorders Branch, Genes, Cognition and Psychosis Program, National Institute of Mental Health, National Institute of Health, National Institutes of Health, Bethesda, MD, USA
| | - Fengyu Zhang
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Heather Decot
- Clinical Brain Disorders Branch, Genes, Cognition and Psychosis Program, National Institute of Mental Health, National Institute of Health, National Institutes of Health, Bethesda, MD, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA,Departments of Psychiatry, Neurology, Neuroscience and the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Amanda J Law
- Departments of Psychiatry and Cell and Developmental Biology, University of Colorado, School of Medicine, Aurora, CO, USA,Departments of Psychiatry and Cell and Developmental Biology, University of Colorado, School of Medicine, Mailstop 8344, RC1 North, RM. 8101, Aurora, CO 80045, USA, Tel: +1 303 724 4418, Fax: +1 303 724 4425, E-mail:
| |
Collapse
|
80
|
Dickinson D, Straub RE, Trampush JW, Gao Y, Feng N, Xie B, Shin JH, Lim HK, Ursini G, Bigos KL, Kolachana B, Hashimoto R, Takeda M, Baum GL, Rujescu D, Callicott JH, Hyde TM, Berman KF, Kleinman JE, Weinberger DR. Differential effects of common variants in SCN2A on general cognitive ability, brain physiology, and messenger RNA expression in schizophrenia cases and control individuals. JAMA Psychiatry 2014; 71:647-56. [PMID: 24718902 PMCID: PMC4160812 DOI: 10.1001/jamapsychiatry.2014.157] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
IMPORTANCE One approach to understanding the genetic complexity of schizophrenia is to study associated behavioral and biological phenotypes that may be more directly linked to genetic variation. OBJECTIVE To identify single-nucleotide polymorphisms associated with general cognitive ability (g) in people with schizophrenia and control individuals. DESIGN, SETTING, AND PARTICIPANTS Genomewide association study, followed by analyses in unaffected siblings and independent schizophrenia samples, functional magnetic resonance imaging studies of brain physiology in vivo, and RNA sequencing in postmortem brain samples. The discovery cohort and unaffected siblings were participants in the National Institute of Mental Health Clinical Brain Disorders Branch schizophrenia genetics studies. Additional schizophrenia cohorts were from psychiatric treatment settings in the United States, Japan, and Germany. The discovery cohort comprised 339 with schizophrenia and 363 community control participants. Follow-up analyses studied 147 unaffected siblings of the schizophrenia cases and independent schizophrenia samples including a total of an additional 668 participants. Imaging analyses included 87 schizophrenia cases and 397 control individuals. Brain tissue samples were available for 64 cases and 61 control individuals. MAIN OUTCOMES AND MEASURES We studied genomewide association with g, by group, in the discovery cohort. We used selected genotypes to test specific associations in unaffected siblings and independent schizophrenia samples. Imaging analyses focused on activation in the prefrontal cortex during working memory. Brain tissue studies yielded messenger RNA expression levels for RefSeq transcripts. RESULTS The schizophrenia discovery cohort showed genomewide-significant association of g with polymorphisms in sodium channel gene SCN2A, accounting for 10.4% of g variance (rs10174400, P = 9.27 × 10(-10)). Control individuals showed a trend for g/genotype association with reversed allelic directionality. The genotype-by-group interaction was also genomewide significant (P = 1.75 × 10(-9)). Siblings showed a genotype association with g parallel to the schizophrenia group and the same interaction pattern. Parallel, but weaker, associations with cognition were found in independent schizophrenia samples. Imaging analyses showed a similar pattern of genotype associations by group and genotype-by-group interaction. Sequencing of RNA in brain revealed reduced expression in 2 of 3 SCN2A alternative transcripts in the patient group, with genotype-by-group interaction, that again paralleled the cognition effects. CONCLUSIONS AND RELEVANCE The findings implicate SCN2A and sodium channel biology in cognitive impairment in schizophrenia cases and unaffected relatives and may facilitate development of cognition-enhancing treatments.
Collapse
Affiliation(s)
- Dwight Dickinson
- Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Richard E. Straub
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Joey W. Trampush
- Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Yuan Gao
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Ningping Feng
- Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Bin Xie
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Hun Ki Lim
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Gianluca Ursini
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
- Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Kristin L. Bigos
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Bhaskar Kolachana
- Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Ryota Hashimoto
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masatoshi Takeda
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Graham L. Baum
- Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Dan Rujescu
- Department of Psychiatry, Ludwig-Maximilians University, Munich, Germany
- Department of Psychiatry, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - Joseph H. Callicott
- Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Thomas M. Hyde
- Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Karen F. Berman
- Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Joel E. Kleinman
- Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Daniel R. Weinberger
- Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
- Departments of Psychiatry, Neurology, Neuroscience and the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
81
|
Mulle JG, Pulver AE, McGrath JA, Wolyniec PS, Dodd AF, Cutler DJ, Sebat J, Malhotra D, Nestadt G, Conrad DF, Hurles M, Barnes CP, Ikeda M, Iwata N, Levinson DF, Gejman PV, Sanders AR, Duan J, Mitchell AA, Peter I, Sklar P, O'Dushlaine CT, Grozeva D, O'Donovan MC, Owen MJ, Hultman CM, Kähler AK, Sullivan PF, Kirov G, Warren ST. Reciprocal duplication of the Williams-Beuren syndrome deletion on chromosome 7q11.23 is associated with schizophrenia. Biol Psychiatry 2014; 75:371-7. [PMID: 23871472 PMCID: PMC3838485 DOI: 10.1016/j.biopsych.2013.05.040] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 05/03/2013] [Accepted: 05/07/2013] [Indexed: 01/10/2023]
Abstract
BACKGROUND Several copy number variants (CNVs) have been implicated as susceptibility factors for schizophrenia (SZ). Some of these same CNVs also increase risk for autism spectrum disorders, suggesting an etiologic overlap between these conditions. Recently, de novo duplications of a region on chromosome 7q11.23 were associated with autism spectrum disorders. The reciprocal deletion of this region causes Williams-Beuren syndrome. METHODS We assayed an Ashkenazi Jewish cohort of 554 SZ cases and 1014 controls for genome-wide CNV. An excess of large rare and de novo CNVs were observed, including a 1.4 Mb duplication on chromosome 7q11.23 identified in two unrelated patients. To test whether this 7q11.23 duplication is also associated with SZ, we obtained data for 14,387 SZ cases and 28,139 controls from seven additional studies with high-resolution genome-wide CNV detection. We performed a meta-analysis, correcting for study population of origin, to assess whether the duplication is associated with SZ. RESULTS We found duplications at 7q11.23 in 11 of 14,387 SZ cases with only 1 in 28,139 control subjects (unadjusted odds ratio 21.52, 95% confidence interval: 3.13-922.6, p value 5.5 × 10(-5); adjusted odds ratio 10.8, 95% confidence interval: 1.46-79.62, p value .007). Of three SZ duplication carriers with detailed retrospective data, all showed social anxiety and language delay premorbid to SZ onset, consistent with both human studies and animal models of the 7q11.23 duplication. CONCLUSIONS We have identified a new CNV associated with SZ. Reciprocal duplication of the Williams-Beuren syndrome deletion at chromosome 7q11.23 confers an approximately tenfold increase in risk for SZ.
Collapse
Affiliation(s)
- Jennifer Gladys Mulle
- Department of Epidemiology, Rollins School of Public Health, Emory University; Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia.
| | - Ann E Pulver
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - John A McGrath
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine
| | - Paula S Wolyniec
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine
| | - Anne F Dodd
- Department of Epidemiology, Rollins School of Public Health, Emory University
| | - David J Cutler
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia
| | - Jonathan Sebat
- Beyster Center for Genomics of Psychiatric Diseases; Department of PsychiatryUniversity of California, San Diego, La Jolla, California; Department of Cellular Molecular and Molecular Medicine, University of California, San Diego, La Jolla, California; Institute for Genomic Medicine, University of California, San Diego, La Jolla, California
| | - Dheeraj Malhotra
- Beyster Center for Genomics of Psychiatric Diseases; Department of PsychiatryUniversity of California, San Diego, La Jolla, California
| | - Gerald Nestadt
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine
| | - Donald F Conrad
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge
| | - Matthew Hurles
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge
| | - Chris P Barnes
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Masashi Ikeda
- Fujita Health University School of Medicine, Toyake, Aichi, Japan
| | - Nakao Iwata
- Fujita Health University School of Medicine, Toyake, Aichi, Japan
| | - Douglas F Levinson
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Pablo V Gejman
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston; Department of Psychiatry and Behavioral Sciences, University of Chicago, Chicago, Illinois
| | - Alan R Sanders
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston; Department of Psychiatry and Behavioral Sciences, University of Chicago, Chicago, Illinois
| | - Jubao Duan
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston; Department of Psychiatry and Behavioral Sciences, University of Chicago, Chicago, Illinois
| | - Adele A Mitchell
- Department of Forensic Biology, Office of Chief Medical Examiner of the City of New York
| | - Inga Peter
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York
| | - Pamela Sklar
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts; Division of Psychiatric Genomics, Department of Psychiatry, Mount Sinai School of Medicine, New York, New York
| | - Colm T O'Dushlaine
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Detelina Grozeva
- Department of Psychological Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael C O'Donovan
- Department of Psychological Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael J Owen
- Department of Psychological Medicine, Cardiff University, Cardiff, United Kingdom
| | - Christina M Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Anna K Kähler
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, North Carolina; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Patrick F Sullivan
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - George Kirov
- Department of Psychological Medicine, Cardiff University, Cardiff, United Kingdom
| | - Stephen T Warren
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia; Departments of Biochemistry and Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
82
|
Jia P, Zhao Z. Network.assisted analysis to prioritize GWAS results: principles, methods and perspectives. Hum Genet 2014; 133:125-38. [PMID: 24122152 PMCID: PMC3943795 DOI: 10.1007/s00439-013-1377-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 10/03/2013] [Indexed: 01/24/2023]
Abstract
Genome-wide association studies (GWAS) have rapidly become a powerful tool in genetic studies of complex diseases and traits. Traditionally, single marker-based tests have been used prevalently in GWAS and have uncovered tens of thousands of disease-associated SNPs. Network-assisted analysis (NAA) of GWAS data is an emerging area in which network-related approaches are developed and utilized to perform advanced analyses of GWAS data in order to study various human diseases or traits. Progress has been made in both methodology development and applications of NAA in GWAS data, and it has already been demonstrated that NAA results may enhance our interpretation and prioritization of candidate genes and markers. Inspired by the strong interest in and high demand for advanced GWAS data analysis, in this review article, we discuss the methodologies and strategies that have been reported for the NAA of GWAS data. Many NAA approaches search for subnetworks and assess the combined effects of multiple genes participating in the resultant subnetworks through a gene set analysis. With no restriction to pre-defined canonical pathways, NAA has the advantage of defining subnetworks with the guidance of the GWAS data under investigation. In addition, some NAA methods prioritize genes from GWAS data based on their interconnections in the reference network. Here, we summarize NAA applications to various diseases and discuss the available options and potential caveats related to their practical usage. Additionally, we provide perspectives regarding this rapidly growing research area.
Collapse
|
83
|
Abstract
The trophic factor neuregulin 1 (Nrg1) and its receptor ErbB4 are schizophrenia candidate genes. NRG1-ErbB4 signaling was thought to regulate spine formation and function in a cell-autonomous manner. Yet, recent studies indicate that ErbB4 expression is largely restricted to GABAergic interneurons and is very low or absent in pyramidal cells. Here, we generated and characterized cell type-specific ErbB4 mutant and transgenic mice. Spine density and the number of excitatory synapses were unaltered by neither deletion nor overexpression of ErbB4 in pyramidal neurons. However, spine density and excitatory synapse number were reduced in PV-ErbB4(-/-) mice where ErbB4 was selectively ablated in parvalbumin-positive GABAergic interneurons. Concurrently, basal glutamate transmission was impaired in PV-ErbB4(-/-) mice, but not in mice where ErbB4 was deleted or overexpressed in pyramidal neurons. Our results demonstrate a role of ErbB4 in PV-positive interneurons for spine formation in excitatory neurons.
Collapse
|
84
|
Gene × environment interaction studies have not properly controlled for potential confounders: the problem and the (simple) solution. Biol Psychiatry 2014; 75:18-24. [PMID: 24135711 PMCID: PMC3859520 DOI: 10.1016/j.biopsych.2013.09.006] [Citation(s) in RCA: 446] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/11/2013] [Accepted: 09/11/2013] [Indexed: 12/15/2022]
Abstract
Candidate gene × environment (G × E) interaction research tests the hypothesis that the effects of some environmental variable (e.g., childhood maltreatment) on some outcome measure (e.g., depression) depend on a particular genetic polymorphism. Because this research is inherently nonexperimental, investigators have been rightly concerned that detected interactions could be driven by confounders (e.g., ethnicity, gender, age, socioeconomic status) rather than by the specified genetic or environmental variables per se. In an attempt to eliminate such alternative explanations for detected G × E interactions, investigators routinely enter the potential confounders as covariates in general linear models. However, this practice does not control for the effects these variables might have on the G × E interaction. Rather, to properly control for confounders, researchers need to enter the covariate × environment and the covariate × gene interaction terms in the same model that tests the G × E term. In this manuscript, I demonstrate this point analytically and show that the practice of improperly controlling for covariates is the norm in the G × E interaction literature to date. Thus, many alternative explanations for G × E findings that investigators had thought were eliminated have not been.
Collapse
|
85
|
McGrath LM, Cornelis MC, Lee PH, Robinson EB, Duncan LE, Barnett JH, Huang J, Gerber G, Sklar P, Sullivan P, Perlis RH, Smoller JW. Genetic predictors of risk and resilience in psychiatric disorders: a cross-disorder genome-wide association study of functional impairment in major depressive disorder, bipolar disorder, and schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2013; 162B:779-88. [PMID: 24039173 PMCID: PMC4019336 DOI: 10.1002/ajmg.b.32190] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 07/09/2013] [Indexed: 01/30/2023]
Abstract
Functional impairment is one of the most enduring, intractable consequences of psychiatric disorders and is both familial and heritable. Previous studies have suggested that variation in functional impairment can be independent of symptom severity. Here we report the first genome-wide association study (GWAS) of functional impairment in the context of major mental illness. Participants of European-American descent (N = 2,246) were included from three large treatment studies of bipolar disorder (STEP-BD) (N = 765), major depressive disorder (STAR*D) (N = 1091), and schizophrenia (CATIE) (N = 390). At study entry, participants completed the SF-12, a widely used measure of health-related quality of life. We performed a GWAS and pathway analysis of the mental and physical components of health-related quality of life across diagnosis (∼1.6 million single nucleotide polymorphisms), adjusting for psychiatric symptom severity. Psychiatric symptom severity was a significant predictor of functional impairment, but it accounted for less than one-third of the variance across disorders. After controlling for diagnostic category and symptom severity, the strongest evidence of genetic association was between variants in ADAMTS16 and physical functioning (P = 5.87 × 10(-8) ). Pathway analysis did not indicate significant enrichment after correction for gene clustering and multiple testing. This study illustrates a phenotypic framework for examining genetic contributions to functional impairment across psychiatric disorders.
Collapse
Affiliation(s)
- Lauren M. McGrath
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | | | - Phil H. Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Elise B. Robinson
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA
| | - Laramie E. Duncan
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA,Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | | | - Jie Huang
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Gloria Gerber
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Pamela Sklar
- Division of Psychiatric Genomics, Mount Sinai School of Medicine, New York, NY
| | - Patrick Sullivan
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Roy H. Perlis
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Jordan W. Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
86
|
Kukshal P, Kodavali VC, Srivastava V, Wood J, McClain L, Bhatia T, Bhagwat AM, Deshpande SN, Nimgaonkar VL, Thelma BK. Dopaminergic gene polymorphisms and cognitive function in a north Indian schizophrenia cohort. J Psychiatr Res 2013; 47:1615-22. [PMID: 23932573 PMCID: PMC3831060 DOI: 10.1016/j.jpsychires.2013.07.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 05/10/2013] [Accepted: 07/05/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Associations of polymorphisms from dopaminergic neurotransmitter pathway genes have mostly been reported in Caucasian ancestry schizophrenia (SZ) samples. As studies investigating single SNPs with SZ have been inconsistent, more detailed analyses utilizing multiple SNPs with the diagnostic phenotype as well as cognitive function may be more informative. Therefore, these analyses were conducted in a north Indian sample. METHODS Indian SZ case-parent trios (n = 601 families); unscreened controls (n = 468) and an independent set of 118 trio families were analyzed. Representative SNPs in the Dopamine D3 receptor (DRD3), dopamine transporter (SLC6A3), vesicular monoamine transporter 2 (SLC18A2), catechol-o-methyltransferase (COMT) and dopamine beta-hydroxylase (DBH) were genotyped using SNaPshot/SNPlex assays (n = 59 SNPs). The Trail Making Test (TMT) was administered to a subset of the sample (n = 260 cases and n = 302 parents). RESULTS Eight SNPs were nominally associated with SZ in either case-control or family based analyses (p < 0.05, rs7631540 and rs2046496 in DRD3; rs363399 and rs10082463 in SLC18A2; rs4680, rs4646315 and rs9332377 in COMT). rs6271 at DBH was associated in both analyses. Haplotypes of DRD3 SNPs incorporating rs7631540-rs2134655-rs3773678-rs324030-rs6280-rs905568 showed suggestive associations in both case-parent and trio samples. At SLC18A2, rs10082463 was nominally associated with psychomotor performance and rs363285 with executive functions using the TMT but did not withstand multiple corrections. CONCLUSIONS Suggestive associations with dopaminergic genes were detected in this study, but convincing links between dopaminergic polymorphisms and SZ or cognitive function were not observed.
Collapse
Affiliation(s)
- Prachi Kukshal
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110 021, India; C. B. Patel Research Centre, Vile Parle (West), Mumbai, India.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Genetics of psychiatric disorders in the GWAS era: an update on schizophrenia. Eur Arch Psychiatry Clin Neurosci 2013; 263 Suppl 2:S147-54. [PMID: 24071914 DOI: 10.1007/s00406-013-0450-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 09/16/2013] [Indexed: 01/21/2023]
Abstract
The influence of genetic factors in the development of schizophrenia has been convincingly demonstrated by family, twin, and adoption studies. The statistical construct of heritability is generally used for estimating the liability due to genetic factors. Heritability estimates for schizophrenia are reported to be between 60 and 80 %. Due to the technical achievements in whole genome-wide association studies, dissection of the underlying genetic factors was intensified recently, resulting in the conclusion that schizophrenia is essentially a polygenic, complex disorder. Most likely more than 100 genes, each with small effect size, contribute to disease risk. A most recent multi-stage genome-wide association study (Ripke et al. in Nat Genet 2013) identified 22 risk loci and estimated that 8,300 independent single-nucleotide polymorphisms contributed to the risk accounting collectively for 32 % in liability. In addition to this polygenic, complex inheritance, there is also strong indication that in some patients a deletion or insertion of a larger chromosomal region [so-called copy number variation (CNV)] might play a crucial role in pathogenesis. This could be specifically important in sporadic cases with schizophrenia, since a higher frequency of de novo mutations has been associated with these CNVs. Further studies, combining much larger sample sizes as well as application of newer technology, such as deep sequencing technologies will be necessary in order to obtain a more comprehensive understanding of the genetic foundations of schizophrenia.
Collapse
|
88
|
Genome-wide association study of schizophrenia using microsatellite markers in the Japanese population. Psychiatr Genet 2013; 23:117-23. [PMID: 23474461 DOI: 10.1097/ypg.0b013e32835fe4f1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVES To search for schizophrenia susceptibility loci, we carried out a case-control study using 28601 microsatellite markers distributed across the entire genome. MATERIALS AND METHODS To control the highly multiple testing, we designed three sequential steps of screening using three independent sets of pooled samples, followed by the confirmatory step using an independent sample set (>2200 case-control pairs). RESULTS The first screening using pooled samples of 157 case-control pairs showed 2966 markers to be significantly associated with the disorder (P<0.05). After the second and the third screening steps using pooled samples of 150 pairs each, 374 markers remained significantly associated with the disorder. We individually genotyped all screening samples using a total of 1536 tag single nucleotide polymorphisms (SNPs) located in the vicinity of ~200 kb from the 59 positive microsatellite markers. Of the 167 SNPs that replicated the significance, we selected 31 SNPs on the basis of the levels of P values for the confirmatory association test using an independent-sample set. The best association signal was observed in rs13404754, located in the upstream region of SLC23A3. We genotyped six additional SNPs in the vicinity of rs13404754. Significant associations were observed in rs13404754, rs6436122, and rs1043160 in the cumulative samples (2617 cases and 2698 controls) (P=0.005, 0.035, and 0.011, respectively). These SNPs are located in the linkage disequilibrium block of 20 kb in size containing SLC23A3, CNPPD1, and FAM134A genes. CONCLUSION Genome-wide association study using microsatellite markers suggested SLC23A3, CNPPD1, and FAM134A genes as candidates for schizophrenia susceptibility in the Japanese population.
Collapse
|
89
|
Walton E, Turner JA, Ehrlich S. Neuroimaging as a potential biomarker to optimize psychiatric research and treatment. Int Rev Psychiatry 2013; 25:619-31. [PMID: 24151806 DOI: 10.3109/09540261.2013.816659] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Complex, polygenic phenotypes in psychiatry hamper our understanding of the underlying molecular pathways and mechanisms of many diseases. The unknown aetiology, together with symptoms which often show a large variability both across individuals and over time and also tend to respond comparatively slowly to medication, can be a problem for patient treatment and drug development. We argue that neuroimaging has the potential to improve psychiatric treatment in two ways. First, by reducing phenotypic complexity, neuroimaging intermediate phenotypes can help to identify disease-related genes and can shed light into the biological mechanisms of known risk genes. Second, quantitative neuroimaging markers - reflecting the spectrum of impairment on a brain-based level - can be used as a more sensitive, reliable and immediate treatment response biomarker. In the end, enhancing both our understanding of the pathophysiology of psychiatric disorders and the prediction of treatment success could eventually optimise current therapy plans.
Collapse
Affiliation(s)
- Esther Walton
- Department of Child and Adolescent Psychiatry, University Hospital Carl Gustav Carus, Dresden University of Technology , Dresden , Germany
| | | | | |
Collapse
|
90
|
Chen WJ. Taiwan Schizophrenia Linkage Study: lessons learned from endophenotype-based genome-wide linkage scans and perspective. Am J Med Genet B Neuropsychiatr Genet 2013; 162B:636-47. [PMID: 24132895 DOI: 10.1002/ajmg.b.32166] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 03/27/2013] [Indexed: 12/26/2022]
Abstract
Taiwan Schizophrenia Linkage Study (TSLS) was initiated with a linkage strategy for locating multiple genes, each of small to moderate effect, and aimed to recruit a large enough sample of pairs of affected siblings and their families ascertained from a multisite study. With a sample of 607 families successfully recruited, a total of 2,242 individuals (1,207 affected and 1,035 unaffected) from 557 families were genotyped using 386 microsatellite markers spaced at an average of 9-cM intervals. Here the author reviews the establishment of TSLS and initial signal derived from linkage scan using the diagnosis of schizophrenia. Based on the limited success of the initial linkage analysis, a sufficient-component causal model is proposed to incorporate endophenotypes and genes for schizophrenia. Four types of candidate endophenotype measured in TSLS, including schizotypal personality, Continuous Performance Test, Wisconsin Card Sorting Test, and niacin skin flush test, are briefly described. The author discusses different strategies of linkage analysis incorporating these endophenotypes, including quantitative trait loci (QTL) linkage analysis, clustering-derived subgroups, ordered subset analysis (OSA), and latent classes for linkage scan. Then the author summarizes the linkage signals generated from seven studies of endophenotype-based linkage analysis using TSLS, including QTL scan of neurocognitive performance, QTL scan of niacin skin flush, the family cluster of attention deficit and execution deficit, OSA by schizophrenia-schizotypy factors, nested OSA by age at onset and neurocognitive performance, and the latent class of deficit schizophrenia for linkage analysis. The perspective of combining next-generation sequencing with linkage analysis of families is also discussed.
Collapse
Affiliation(s)
- Wei J Chen
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Genetic Epidemiology Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan; Department of Psychiatry, College of Medicine and National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan; Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
91
|
MESH Headings
- Causality
- Chromosome Breakpoints
- Chromosomes, Human, Pair 1/genetics
- Chromosomes, Human, Pair 1/ultrastructure
- Chromosomes, Human, Pair 11/genetics
- Chromosomes, Human, Pair 11/ultrastructure
- Conduct Disorder/genetics
- Genetic Predisposition to Disease
- Genome-Wide Association Study
- Humans
- Lod Score
- Mental Disorders/genetics
- Mood Disorders/genetics
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/physiology
- Pedigree
- Phenotype
- RNA, Long Noncoding/genetics
- Risk Factors
- Schizophrenia/genetics
- Terminology as Topic
- Translocation, Genetic
Collapse
Affiliation(s)
- P F Sullivan
- Departments of Genetics and Psychiatry, Center for Psychiatric Genomics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
92
|
Variants in the 15q25 gene cluster are associated with risk for schizophrenia and bipolar disorder. Psychiatr Genet 2013. [PMID: 23196875 DOI: 10.1097/ypg.0b013e32835bd5f1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Rates of tobacco smoking are significantly higher in patients with schizophrenia compared with the general population. The underlying mechanism for this comorbidity is unclear. One hypothesis is that there are common genetic factors that predispose to both nicotine dependence (ND) and schizophrenia. To investigate this hypothesis, we examined the association of the 15q25 gene cluster, the most significant candidate region to date implicated in ND and smoking behavior, with schizophrenia and bipolar disorder. METHODS Five variants in the 15q25 gene cluster (rs951266, rs16969968, rs1051730, rs8040868, and rs17477223) were selected to test for association with schizophrenia diagnosis, bipolar disorder diagnosis, and the presence of negative symptoms of schizophrenia. Effects of the variants on 15q25 gene expression were analyzed using publically available postmortem brain expression data. RESULTS A meta-analysis revealed four markers associated with risk for schizophrenia and bipolar disorder (rs951266, rs16969968, rs8040868, and rs17477223), and with the presence of negative symptoms of schizophrenia (rs951266, rs1051730, rs8040868, and rs17477223). The associations were in the same direction as that found for ND. Gene expression analysis indicated an association between genotypes of the rs1051730 variant and CHRNA5 expression in brain and peripheral blood mononuclear cells, and with the rs16969968 and rs17477223 variants in brain. CONCLUSION Variants in the 15q25 gene cluster are associated with risk for schizophrenia/bipolar illness, negative symptoms of schizophrenia, and influence CHRNA5 expression in the brain and peripheral blood mononuclear cells. These results are consistent with the notion that there are genetic mechanisms common to schizophrenia, ND, and bipolar disorder.
Collapse
|
93
|
Murphy E, McMahon FJ. Pharmacogenetics of antidepressants, mood stabilizers, and antipsychotics in diverse human populations. DISCOVERY MEDICINE 2013; 16:113-122. [PMID: 23998447 PMCID: PMC6011657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
An increasing focus on personalized medicine is driving a renewed effort to understand the impact of ethnic and genetic background on treatment outcomes. Since responses to psychopharmacological treatments continue to be sub-optimal, there is a pressing need to identify markers of tolerability and efficacy. Pharmacogenomic studies aim to find such markers within the human genome, and have made some progress in recent years. Progress has been slower in populations with diverse racial and ethnic backgrounds. Here we review 10 genome-wide association studies (GWAS) that assessed outcomes after antidepressant, antipsychotic, or mood stabilizer treatment. These studies used samples collected by the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE), Sequenced Treatment Alternatives to Relieve Depression (STAR*D), and Systematic Treatment Enhancement Program for Bipolar Disorder (STEP-BD) studies. We highlight findings from African American and European American participants since they are the largest groups studied, but we also address issues related to Asian and Hispanic groups. None of the GWAS we reviewed identified individual genetic markers at genome-wide significance, probably due to limited sample sizes. However, all the studies found poorer outcomes among African American participants. Some of this disparity seems to be explained by psychosocial and economic disadvantages, but at least 2 studies found that widespread genetic differences between participants of European and African ancestry also play an important role. Non-European groups are underrepresented in these studies, but the differences that are evident so far suggest that poorer outcomes among African Americans are not inevitable and may be particularly suited to pharmacogenomic strategies. The vision of more personalized psychopharmacology may critically depend on larger studies in more diverse human populations.
Collapse
Affiliation(s)
- Eleanor Murphy
- Human Genetics Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
94
|
Evidence for shared genetic risk between methamphetamine-induced psychosis and schizophrenia. Neuropsychopharmacology 2013; 38:1864-70. [PMID: 23594818 PMCID: PMC3746703 DOI: 10.1038/npp.2013.94] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 03/25/2013] [Accepted: 03/26/2013] [Indexed: 02/04/2023]
Abstract
Methamphetamine (METH) use can provoke psychotic reactions requiring immediate treatment, namely METH-induced psychosis. Although the distinction between METH-induced and primary psychosis is important for understanding their clinical courses, we do not have clear diagnostic procedure by their symptoms. Not only are there similarities between the clinical features of METH-induced psychosis and schizophrenia (SCZ), but there is also epidemiological evidence of a shared genetic risk between 'METH-related' disorders and SCZ, which makes the differentiation of these two conditions difficult. In this study, we conducted a genome-wide association study (GWAS) targeting METH-dependent patients. The METH sample group, used in the METH-dependence GWAS, included 236 METH-dependent patients and 864 healthy controls. We also included a 'within-case' comparison between 194 METH-induced psychosis patients and 42 METH-dependent patients without psychosis in a METH-induced psychosis GWAS. To investigate the shared genetic components between METH dependence, METH-induced psychosis, and SCZ, data from our previous SCZ GWAS (total N=1108) were re-analyzed. In the SNP-based analysis, none of the SNPs showed genome-wide significance in either data set. By performing a polygenic component analysis, however, we found that a large number of 'risk' alleles for METH-induced psychosis are over-represented in individuals with SCZ (Pbest=0.0090). Conversely, we did not detect enrichment either between METH dependence and METH-induced psychosis or between METH dependence and SCZ. The results support previous epidemiological and neurobiological evidence for a relationship between METH-induced psychosis and SCZ. These also suggest that the overlap between genes scored as positive in these data sets can have higher probability as susceptibility genes for psychosis.
Collapse
|
95
|
Ramsey TL, Liu Q, Massey BW, Brennan MD. Genotypic variation in the SV2C gene impacts response to atypical antipsychotics the CATIE study. Schizophr Res 2013; 149:21-5. [PMID: 23886675 PMCID: PMC3845218 DOI: 10.1016/j.schres.2013.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/27/2013] [Accepted: 07/02/2013] [Indexed: 11/27/2022]
Abstract
Pharmacogenetic (PGx) predictors of response would improve outcomes in antipsychotic treatment. Based on both biological rationale and prior evidence of an impact on Parkinson's disease, we conducted an association study for 106 SNPs in the synaptic vesicle protein 2C (SV2C) gene using genetic and treatment response data from the Clinical Trial of Antipsychotic Intervention Effectiveness (CATIE). We examined response to the atypical antipsychotics for Caucasian subjects in the blinded phases, Phases 1A, 1B, and 2, of CATIE with sample sizes as follows: olanzapine (N=134), quetiapine (N=124), risperidone (N=134), and ziprasidone (N=74). Response was defined as change in the Positive and Negative Syndrome Scale (PANSS) score using a mixed model repeat measures approach. Subjects homozygous for the T allele of rs11960832 displayed significantly worse response to olanzapine treatment, the only finding with study-wide significance (p=2.94×10(-5); false discovery rate=2.18×10(-2)). These subjects also displayed worse response to quetiapine with nominal significance (p=4.56×10(-2)). While no other SNP achieved study-wide significance, one SNP (rs10214163) influencing Parkinson's disease displayed nominally significant association with olanzapine and quetiapine response, while the second such SNP (rs30196) showed a statistical trend toward correlating with olanzapine and quetiapine response. Furthermore, both coding SNPs examined (rs31244 and rs2270927) displayed nominally significant correlations with treatment response: one for olanzapine (rs227092), and one for quetiapine (rs31244). The fact that multiple SNPs in SV2C may impact response to atypical antipsychotics suggests that further evaluation of SNPs in this gene as PGx predictors of antipsychotic response is warranted.
Collapse
Affiliation(s)
- Timothy L. Ramsey
- SureGene, LLC, 600 Envoy Circle, suite 601, Louisville, KY 40299 United States
| | - Qian Liu
- SureGene, LLC, 600 Envoy Circle, suite 601, Louisville, KY 40299 United States
| | - Bill W. Massey
- SureGene, LLC, 600 Envoy Circle, suite 601, Louisville, KY 40299 United States
| | - Mark D. Brennan
- SureGene, LLC, 600 Envoy Circle, suite 601, Louisville, KY 40299 United States,Communicating author, Mark D. Brennan, SureGene, LLC, 600 Envoy Circle, suite 601, Louisville, KY 40299 United States, , Phone: 502-287-0899, Fax: 859-663-2984
| |
Collapse
|
96
|
Zhao Z, Webb BT, Jia P, Bigdeli TB, Maher BS, van den Oord E, Bergen SE, Amdur RL, O'Neill FA, Walsh D, Thiselton DL, Chen X, Pato CN, The International Schizophrenia Consortium, Riley BP, Kendler KS, Fanous AH. Association study of 167 candidate genes for schizophrenia selected by a multi-domain evidence-based prioritization algorithm and neurodevelopmental hypothesis. PLoS One 2013; 8:e67776. [PMID: 23922650 PMCID: PMC3726675 DOI: 10.1371/journal.pone.0067776] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 05/28/2013] [Indexed: 01/06/2023] Open
Abstract
Integrating evidence from multiple domains is useful in prioritizing disease candidate genes for subsequent testing. We ranked all known human genes (n = 3819) under linkage peaks in the Irish Study of High-Density Schizophrenia Families using three different evidence domains: 1) a meta-analysis of microarray gene expression results using the Stanley Brain collection, 2) a schizophrenia protein-protein interaction network, and 3) a systematic literature search. Each gene was assigned a domain-specific p-value and ranked after evaluating the evidence within each domain. For comparison to this ranking process, a large-scale candidate gene hypothesis was also tested by including genes with Gene Ontology terms related to neurodevelopment. Subsequently, genotypes of 3725 SNPs in 167 genes from a custom Illumina iSelect array were used to evaluate the top ranked vs. hypothesis selected genes. Seventy-three genes were both highly ranked and involved in neurodevelopment (category 1) while 42 and 52 genes were exclusive to neurodevelopment (category 2) or highly ranked (category 3), respectively. The most significant associations were observed in genes PRKG1, PRKCE, and CNTN4 but no individual SNPs were significant after correction for multiple testing. Comparison of the approaches showed an excess of significant tests using the hypothesis-driven neurodevelopment category. Random selection of similar sized genes from two independent genome-wide association studies (GWAS) of schizophrenia showed the excess was unlikely by chance. In a further meta-analysis of three GWAS datasets, four candidate SNPs reached nominal significance. Although gene ranking using integrated sources of prior information did not enrich for significant results in the current experiment, gene selection using an a priori hypothesis (neurodevelopment) was superior to random selection. As such, further development of gene ranking strategies using more carefully selected sources of information is warranted.
Collapse
Affiliation(s)
- Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Bradley T. Webb
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Center for Biomarker Research and Personalized Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Peilin Jia
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - T. Bernard Bigdeli
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Brion S. Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Edwin van den Oord
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Center for Biomarker Research and Personalized Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Sarah E. Bergen
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Richard L. Amdur
- Washington VA Medical Center, Washington, DC, United States of America
| | | | | | - Dawn L. Thiselton
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Xiangning Chen
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Carlos N. Pato
- Department of Psychiatry, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | | | - Brien P. Riley
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Ayman H. Fanous
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Washington VA Medical Center, Washington, DC, United States of America
- Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychiatry, Georgetown University School of Medicine, Washington, DC, United States of America
- Department of Psychiatry, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
97
|
Green MJ, Cairns MJ, Wu J, Dragovic M, Jablensky A, Tooney PA, Scott RJ, Carr VJ. Genome-wide supported variant MIR137 and severe negative symptoms predict membership of an impaired cognitive subtype of schizophrenia. Mol Psychiatry 2013; 18:774-80. [PMID: 22733126 DOI: 10.1038/mp.2012.84] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Progress in determining the aetiology of schizophrenia (Sz) has arguably been limited by a poorly defined phenotype. We sought to delineate empirically derived cognitive subtypes of Sz to investigate the association of a genetic variant identified in a recent genome-wide association study with specific phenotypic characteristics of Sz. We applied Grade of Membership (GoM) analyses to 617 patients meeting ICD-10 criteria for Sz (n=526) or schizoaffective disorder (n=91), using cognitive performance indicators collected within the Australian Schizophrenia Research Bank. Cognitive variables included subscales from the Repeatable Battery for the Assessment of Neuropsychological Status, the Controlled Oral Word Association Test and the Letter Number Sequencing Test, and standardised estimates of premorbid and current intelligence quotient. The most parsimonious GoM solution yielded two subtypes of clinical cases reflecting those with cognitive deficits (CDs; N=294), comprising 47.6% of the sample who were impaired across all cognitive measures, and a cognitively spared group (CS; N=323) made up of the remaining 52.4% who performed relatively well on all cognitive tests. The CD subgroup were more likely to be unemployed, had an earlier illness onset, and greater severity of functional disability and negative symptoms than the CS group. Risk alleles on the MIR137 single-nucleotide polymorphism (SNP) predicted membership of CD subtype only in combination with higher severity of negative symptoms. These findings provide the first evidence for association of the MIR137 SNP with a specific Sz phenotype characterised by severe CDs and negative symptoms, consistent with the emerging role of microRNAs in the regulation of proteins responsible for neural development and function.
Collapse
Affiliation(s)
- M J Green
- School of Psychiatry, University of New South Wales, Research Unit for Schizophrenia Epidemiology, St Vincent's Hospital, Darlinghurst, NSW, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Genome-wide association study of patient-rated and clinician-rated global impression of severity during antipsychotic treatment. Pharmacogenet Genomics 2013; 23:69-77. [PMID: 23241943 DOI: 10.1097/fpc.0b013e32835ca260] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To examine the unique and congruent findings between multiple raters in a genome-wide association study (GWAS) in the context of understanding individual differences in treatment response during antipsychotic therapy for schizophrenia. MATERIALS AND METHODS We performed GWAS to search for genetic variation affecting treatment response. The analysis sample included 738 patients with schizophrenia, successfully genotyped for ∼492k single nucleotide polymorphisms (SNPs) from the Clinical Antipsychotic Trial of Intervention Effectiveness. Outcomes included both clinician and patient report of illness severity on global impression scales, the clinical global impression severity scale and patient global impression, respectively. Our criterion for genome-wide significance was a prespecified threshold ensuring that, on average, only 10% of the significant findings are false discoveries. RESULTS Thirteen SNPs reached genome-wide significance. The top findings indicated three SNPs in PDE4D, 5q12.1 (P=4.2×10, 1.6×10, 1.8×10), mediating the effects of quetiapine on patient-reported severity and an additional three SNPs in TJP1, 15q13.1 (P=2.25×10, 4.86×10, 4.91×10), mediating the effects of risperidone on patient-reported severity. For clinician-reported severity, two SNPs in PPA2, 4q24 (P=3.68×10, 5.05×10), were found to reach genome-wide significance. CONCLUSION We found evidence of both a novel and a consistent association when examining the results from the patient and clinician ratings, suggesting that different raters may capture unique facets of schizophrenia. Although our findings require replication and functional validation, this study shows the potential of GWAS to discover genes that potentially mediate treatment response of antipsychotic medication.
Collapse
|
99
|
Guan F, Wei S, Zhang C, Zhang H, Zhang B, Xing B, Feng Z, Gao C, Liu X, Li S. A population-based association study of 2q32.3 and 8q21.3 loci with schizophrenia in Han Chinese. J Psychiatr Res 2013; 47:712-717. [PMID: 23490065 DOI: 10.1016/j.jpsychires.2013.01.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Revised: 01/29/2013] [Accepted: 01/29/2013] [Indexed: 12/21/2022]
Abstract
It has been reported that two new schizophrenia susceptibility loci (2q32.3 and 8q21.3) containing two single-nucleotide polymorphisms (SNPs; rs17662626 and rs7004633) have been identified in Europeans by a genome-wide association study. To determine if the two regions are associated with schizophrenia in Han Chinese, which are distinct from Europeans, we analyzed 9 SNPs, including rs17662626 and rs7004633, within 2 regions involving 1430 cases and 1570 controls from the Han population. Single SNP association, haplotype association, and gender-specific association analyses were performed. Single SNP analyses revealed that there was no association with schizophrenia in the region of 2q32.3, but rs7004633 mapping to the region of 8q21.3 was significantly associated with schizophrenia (p = 5.1 × 10(-5); OR = 1.274; 95% CI, 1.134-1.429). Further genotype and haplotype association analyses for the region of 8q21.3 suggested a similar pattern. Additionally, analyses by haplotypes indicated that a haplotype block in the region of 8q21.3 highly associated with schizophrenia and one haplotype in this haploblock had a 1.5-fold increase in the cases. Our results provide further evidence regarding the association of the region of 8q21.3 with schizophrenia in Han Chinese, as well as Europeans, which confirmed the previous report that 8q21.3 may play important roles in the etiology of schizophrenia.
Collapse
Affiliation(s)
- Fanglin Guan
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Weißflog L, Scholz CJ, Jacob CP, Nguyen TT, Zamzow K, Groß-Lesch S, Renner TJ, Romanos M, Rujescu D, Walitza S, Kneitz S, Lesch KP, Reif A. KCNIP4 as a candidate gene for personality disorders and adult ADHD. Eur Neuropsychopharmacol 2013; 23:436-47. [PMID: 22981920 DOI: 10.1016/j.euroneuro.2012.07.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 07/06/2012] [Accepted: 07/26/2012] [Indexed: 11/19/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder in children with striking persistence into adulthood and a high co-morbidity with other psychiatric disorders, including personality disorders (PD). The 4p15.31 region was shown to be associated with ADHD in several genome wide association studies (GWAS). In the present study we also report association of the 4p15.31 locus with Cluster B and Cluster C PD as identified by a pooled genome-wide association study in 400 individuals suffering from PD. The gene coding for the Kv channel-interacting protein 4 (KCNIP4) is located in this region. KCNIP4 is an interaction partner of presenilin and plays a role in a negative feedback loop in the Wnt/β-catenin pathway. Thus, we reasoned it to be a promising candidate gene for ADHD as well as for PD. To clarify the role of KCNIP4 in those disorders, we conducted candidate gene based association studies in 594 patients suffering from adult ADHD and 630 PD patients as compared to 974 healthy control individuals. In the adult ADHD sample, six single markers and one haplotype block revealed to be associated with disease (p values from 0.0079 to 0.049). Seven markers within the KCNIP4 gene showed an association with PD (p values from 0.0043 to 0.0437). The results of these studies suggest a role of KCNIP4 in the etiology of ADHD, PD and other co-morbid disorders.
Collapse
Affiliation(s)
- Lena Weißflog
- Department of Psychiatry, ADHD Clinical Research Network, Molecular Psychiatry Laboratory of Translational Neuroscience; Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|