101
|
Tamariz L, Balda J, Pareja D, Palacio A, Myerburg RJ, Conway D, Davis L, Goldberger JJ. Usefulness of Single Nucleotide Polymorphisms as Predictors of Sudden Cardiac Death. Am J Cardiol 2019; 123:1900-1905. [PMID: 31053292 DOI: 10.1016/j.amjcard.2019.02.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/23/2022]
Abstract
The pathophysiology of sudden cardiac death (SCD) remains incompletely understood. Genetic mutations can create a favorable substrate for SCD. Our aim is to evaluate the evidence of single nucleotide polymorphisms (SNPs) as predictors of SCD. We searched the Medline database (2000 to 2017) and selected all case-control or cohort studies that reported associations between SNPs and SCD. Our search terms included "polymorphisms" and "sudden death." We collected the study design, population ethnic background, gene testing strategy, the association between the SNP and SCD, and the cardiovascular comorbidities of the population. Our search yielded 723 studies, of which we included 24 based upon our inclusion criteria. The studies had a total population of 78,165 participants, with a median age of 62.5 years (IQR 56 to 66) and 35% (IQR 13 to 32) were female. Almost all studies were conducted in white patients of European descent and the most commonly used genetic strategy was candidate gene panels. Fifteen of the studies had a case-control design that included SCD patients without known heart disease as the comparison group and the other 9 studies included patients with heart failure and coronary artery disease. The studies evaluated 53 SNPs and the most common genetic loci were SCN5A, RyR2, CASQ2, NOSA1P, and AGTR. SNPs with the 3 strongest statistically significant ORs >1 were: rs6684209 of CASQ2 (odds ratio [OR] 19), rs3814843 of CALM1 (OR 5.5), and rs35594137 of GJA5 (OR 3.6). In Conclusion, many SNPs are associated with SCD, with the strongest associations seen in SNPs of genes related to intracellular calcium handling. These findings were generated primarily using a candidate gene strategy in white patients with European descent.
Collapse
Affiliation(s)
- Leonardo Tamariz
- Division of Population Health and Computational Medicine, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida; the Geriatric Research Education and Clinical Center, Veterans Affairs Medical Center, Miami, Florida.
| | - Javier Balda
- Department of Medicine, St Elizabeths Medical Center, Boston, Massachusetts; Universidad Catolica Santiago de Guayaquil, Guayaquil, Ecuador
| | - Dennise Pareja
- the Geriatric Research Education and Clinical Center, Veterans Affairs Medical Center, Miami, Florida; Universidad Catolica Santiago de Guayaquil, Guayaquil, Ecuador
| | - Ana Palacio
- Division of Population Health and Computational Medicine, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida; the Geriatric Research Education and Clinical Center, Veterans Affairs Medical Center, Miami, Florida
| | - Robert J Myerburg
- Division of Cardiology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| | - Douglas Conway
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee
| | - Lea Davis
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee
| | - Jeffrey J Goldberger
- Division of Cardiology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
102
|
Chellappa SA, Pathak AK, Sinha P, Jainarayanan ASHWINK, Jain S, Brahmachari SK. Meta-analysis of genomic variants and gene expression data in schizophrenia suggests the potential need for adjunctive therapeutic interventions for neuropsychiatric disorders. J Genet 2019. [DOI: 10.1007/s12041-019-1101-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
103
|
Anirudh Chellappa S, Pathak AK, Sinha P, Jainarayanan AK, Jain S, Brahmachari SK. Meta-analysis of genomic variants and gene expression data in schizophrenia suggests the potential need for adjunctive therapeutic interventions for neuropsychiatric disorders. J Genet 2019; 98:60. [PMID: 31204709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Schizophrenia (SZ) is a debilitating mental illness with a multigenic aetiology and significant heritability. Despite extensive genetic studies, the molecular aetiology has remained enigmatic. A recent systems biology study suggested a protein-protein interaction network for SZ with 504 novel interactions. The onset of psychiatric disorders is predominant during adolescence, often accompanied by subtle structural abnormalities in multiple regions of the brain. The availability of BrainSpan Atlas data allowed us to re-examine the genes present in the SZ interactome as a function of space and time. The availability of genomes of healthy centenarians and nonpsychiatric Exome Aggregation Consortium database allowed us to identify the variants of criticality. The expression of the SZ candidate genes responsible for cognition and disease onset was studied in different brain regions during particular developmental stages. A subset of novel interactors detected in the network was further validated using gene expression data of post-mortem brains of patients with psychiatric illness. We have narrowed down the list of drug targets proposed by theprevious interactome study to 10 proteins. These proteins belonging to 81 biological pathways are targeted by 34 known Food and Drug Administration-approved drugs that have distinct potential for the treatment of neuropsychiatric disorders. We also report the possibility of targeting key genes belonging to celecoxib pharmacodynamics, Gα signalling and cGMP-PKG signalling pathwaysthat are not known to be specific to SZ aetiology.
Collapse
Affiliation(s)
- S Anirudh Chellappa
- Centre for Open Innovation - Indian Centre for Social Transformation (ICST), Bengaluru 560 001, India
| | | | | | | | | | | |
Collapse
|
104
|
Dezhina Z, Ranlund S, Kyriakopoulos M, Williams SCR, Dima D. A systematic review of associations between functional MRI activity and polygenic risk for schizophrenia and bipolar disorder. Brain Imaging Behav 2019; 13:862-877. [PMID: 29748770 PMCID: PMC6538577 DOI: 10.1007/s11682-018-9879-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Genetic factors account for up to 80% of the liability for schizophrenia (SCZ) and bipolar disorder (BD). Genome-wide association studies have successfully identified several genes associated with increased risk for both disorders. This has allowed researchers to model the aggregate effect of genes associated with disease status and create a polygenic risk score (PGRS) for each individual. The interest in imaging genetics using PGRS has grown in recent years, with several studies now published. We have conducted a systematic review to examine the effects of PGRS of SCZ, BD and cross psychiatric disorders on brain function and connectivity using fMRI data. Results indicate that the effect of genetic load for SCZ and BD on brain function affects task-related recruitment, with frontal areas having a more prominent role, independent of task. Additionally, the results suggest that the polygenic architecture of psychotic disorders is not regionally confined but impacts on the task-dependent recruitment of multiple brain regions. Future imaging genetics studies with large samples, especially population studies, would be uniquely informative in mapping the spatial distribution of the genetic risk to psychiatric disorders on brain processes during various cognitive tasks and may lead to the discovery of biological pathways that could be crucial in mediating the link between genetic factors and alterations in brain networks.
Collapse
Affiliation(s)
- Zalina Dezhina
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Siri Ranlund
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Marinos Kyriakopoulos
- National and Specialist Acorn Lodge Inpatient Children Unit, South London and Maudsley NHS Foundation Trust, London, UK
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Steve C R Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Danai Dima
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- Department of Psychology, School of Arts and Social Sciences, City, University of London, 10 Northampton Square, London, EC1V 0HB, UK.
| |
Collapse
|
105
|
Jouroukhin Y, Zhu X, Shevelkin AV, Hasegawa Y, Abazyan B, Saito A, Pevsner J, Kamiya A, Pletnikov MV. Adolescent Δ 9-Tetrahydrocannabinol Exposure and Astrocyte-Specific Genetic Vulnerability Converge on Nuclear Factor-κB-Cyclooxygenase-2 Signaling to Impair Memory in Adulthood. Biol Psychiatry 2019; 85:891-903. [PMID: 30219209 PMCID: PMC6525084 DOI: 10.1016/j.biopsych.2018.07.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/19/2018] [Accepted: 07/30/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND Although several studies have linked adolescent cannabis use to long-term cognitive dysfunction, there are negative reports as well. The fact that not all users develop cognitive impairment suggests a genetic vulnerability to adverse effects of cannabis, which are attributed to action of Δ9-tetrahydrocannabinol (Δ9-THC), a cannabis constituent and partial agonist of brain cannabinoid receptor 1. As both neurons and glial cells express cannabinoid receptor 1, genetic vulnerability could influence Δ9-THC-induced signaling in a cell type-specific manner. METHODS Here we use an animal model of inducible expression of dominant-negative disrupted in schizophrenia 1 (DN-DISC1) selectively in astrocytes to evaluate the molecular mechanisms, whereby an astrocyte genetic vulnerability could interact with adolescent Δ9-THC exposure to impair recognition memory in adulthood. RESULTS Selective expression of DN-DISC1 in astrocytes and adolescent treatment with Δ9-THC synergistically affected recognition memory in adult mice. Similar deficits in recognition memory were observed following knockdown of endogenous Disc1 in hippocampal astrocytes in mice treated with Δ9-THC during adolescence. At the molecular level, DN-DISC1 and Δ9-THC synergistically activated the nuclear factor-κB-cyclooxygenase-2 pathway in astrocytes and decreased immunoreactivity of parvalbumin-positive presynaptic inhibitory boutons around pyramidal neurons of the hippocampal CA3 area. The cognitive abnormalities were prevented in DN-DISC1 mice exposed to Δ9-THC by simultaneous adolescent treatment with the cyclooxygenase-2 inhibitor, NS398. CONCLUSIONS Our data demonstrate that individual vulnerability to cannabis can be exclusively mediated by astrocytes. Results of this work suggest that genetic predisposition within astrocytes can exaggerate Δ9-THC-produced cognitive impairments via convergent inflammatory signaling, suggesting possible targets for preventing adverse effects of cannabis within susceptible individuals.
Collapse
Affiliation(s)
- Yan Jouroukhin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xiaolei Zhu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexey V Shevelkin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yuto Hasegawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bagrat Abazyan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Atsushi Saito
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan Pevsner
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland; Kennedy Krieger Institute, Baltimore, Maryland
| | - Atsushi Kamiya
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
106
|
Border R, Johnson EC, Evans LM, Smolen A, Berley N, Sullivan PF, Keller MC. No Support for Historical Candidate Gene or Candidate Gene-by-Interaction Hypotheses for Major Depression Across Multiple Large Samples. Am J Psychiatry 2019; 176:376-387. [PMID: 30845820 PMCID: PMC6548317 DOI: 10.1176/appi.ajp.2018.18070881] [Citation(s) in RCA: 334] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Interest in candidate gene and candidate gene-by-environment interaction hypotheses regarding major depressive disorder remains strong despite controversy surrounding the validity of previous findings. In response to this controversy, the present investigation empirically identified 18 candidate genes for depression that have been studied 10 or more times and examined evidence for their relevance to depression phenotypes. METHODS Utilizing data from large population-based and case-control samples (Ns ranging from 62,138 to 443,264 across subsamples), the authors conducted a series of preregistered analyses examining candidate gene polymorphism main effects, polymorphism-by-environment interactions, and gene-level effects across a number of operational definitions of depression (e.g., lifetime diagnosis, current severity, episode recurrence) and environmental moderators (e.g., sexual or physical abuse during childhood, socioeconomic adversity). RESULTS No clear evidence was found for any candidate gene polymorphism associations with depression phenotypes or any polymorphism-by-environment moderator effects. As a set, depression candidate genes were no more associated with depression phenotypes than noncandidate genes. The authors demonstrate that phenotypic measurement error is unlikely to account for these null findings. CONCLUSIONS The study results do not support previous depression candidate gene findings, in which large genetic effects are frequently reported in samples orders of magnitude smaller than those examined here. Instead, the results suggest that early hypotheses about depression candidate genes were incorrect and that the large number of associations reported in the depression candidate gene literature are likely to be false positives.
Collapse
Affiliation(s)
- Richard Border
- Institute for Behavioral Genetics, University of Colorado Boulder, Stockholm, Sweden,Department of Psychology and Neuroscience, University of Colorado Boulder, Stockholm, Sweden,Department of Applied Mathematics, University of Colorado Boulder, Stockholm, Sweden
| | - Emma C. Johnson
- Institute for Behavioral Genetics, University of Colorado Boulder, Stockholm, Sweden,Department of Psychiatry, Washington University School of Medicine, Stockholm, Sweden
| | - Luke M. Evans
- Institute for Behavioral Genetics, University of Colorado Boulder, Stockholm, Sweden,Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Stockholm, Sweden
| | - Andrew Smolen
- Institute for Behavioral Genetics, University of Colorado Boulder, Stockholm, Sweden
| | - Noah Berley
- Department of Psychology and Neuroscience, University of Colorado Boulder, Stockholm, Sweden
| | - Patrick F. Sullivan
- Department of Genetics and Psychiatry, University of North Carolina at Chapel Hill, Stockholm, Sweden,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Matthew C. Keller
- Institute for Behavioral Genetics, University of Colorado Boulder, Stockholm, Sweden,Department of Psychology and Neuroscience, University of Colorado Boulder, Stockholm, Sweden
| |
Collapse
|
107
|
Dick DM. Commentary for Special Issue of Prevention Science "Using Genetics in Prevention: Science Fiction or Science Fact?". PREVENTION SCIENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR PREVENTION RESEARCH 2019; 19:101-108. [PMID: 28735446 DOI: 10.1007/s11121-017-0828-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A growing number of prevention studies have incorporated genetic information. In this commentary, I discuss likely reasons for growing interest in this line of research and reflect on the current state of the literature. I review challenges associated with the incorporation of genotypic information into prevention studies, as well as ethical considerations associated with this line of research. I discuss areas where developmental psychologists and prevention scientists can make substantive contributions to the study of genetic predispositions, as well as areas that could benefit from closer collaborations between prevention scientists and geneticists to advance this area of study. In short, this commentary tackles the complex questions associated with what we hope to achieve by adding genetic components to prevention research and where this research is likely to lead in the future.
Collapse
Affiliation(s)
- Danielle M Dick
- Departments of Psychology and Human & Molecular Genetics, College Behavioral and Emotional Health Institute, Virginia Commonwealth University, Box 842018, Richmond, VA, 23284-2018, USA.
| |
Collapse
|
108
|
Female gender specific association of the Reelin (RELN) gene rs7341475 variant with schizophrenia. Mol Biol Rep 2019; 46:3411-3416. [PMID: 30980267 DOI: 10.1007/s11033-019-04803-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 04/05/2019] [Indexed: 12/25/2022]
Abstract
RELN gene encodes a large extracellular matrix protein which is critical for neuronal migration, cell positioning and cell-cell interactions. It also controls the synaptic plasticity of neurons for initiation and maintenance of long term potentiation. The aim of this study is to investigate the association of RELN rs7341475 variant with schizophrenia. Genomic DNA isolation was performed from 105 schizophrenic patients and 137 healthy controls to determine RELN rs7341475 genotypes. Genotype and allele frequencies were determined by a polymerase chain reaction-restriction fragment length polymorphism method developed in our laboratory. Statistical analysis was performed using χ2 test. The frequencies for G allele were 79.5% in cases and 81.0% in controls, for A allele 20.5% in cases and 19.0% in controls in the overall population. The genotype frequencies of the RELN gene rs7341475 variant were GG; 63.8%, GA; 31.4% and AA; 4.8% in cases, GG; 63.5%, GA; 35.0% and AA; 1.5% in controls in the overall population. There was no statistically significant association between the rs7341475 variant of RELN gene and schizophrenia in the overall population (χ2 = 2.473, p = 0.290). In the gender specific analysis, female gender specific association was only found. The RELN rs7341475 variant GG genotype was significantly associated with schizophrenia (p = 0.034, OR 2.760, 95% CI 1.058-7.197) and A allele was protective against schizophrenia (p = 0.034, OR 0.362, 95% CI 0.139-0.945). All cases and controls were in Hardy-Weinberg equilibrium (p > 0.05). Population size can be increased to improve the statistical power. Moreover, other RELN gene variants which are especially involved in neuronal migration and epigenetic regulation may be analyzed for revealing the complex genetic architecture of schizophrenia. In conclusion, there was only association between the RELN rs7341475 variant and schizophrenia in the female gender in a Turkish population.
Collapse
|
109
|
McIntosh AM, Sullivan PF, Lewis CM. Uncovering the Genetic Architecture of Major Depression. Neuron 2019; 102:91-103. [PMID: 30946830 PMCID: PMC6482287 DOI: 10.1016/j.neuron.2019.03.022] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/05/2019] [Accepted: 03/14/2019] [Indexed: 12/21/2022]
Abstract
There have been several recent studies addressing the genetic architecture of depression. This review serves to take stock of what is known now about the genetics of depression, how it has increased our knowledge and understanding of its mechanisms, and how the information and knowledge can be leveraged to improve the care of people affected. We identify four priorities for how the field of MD genetics research may move forward in future years, namely by increasing the sample sizes available for genome-wide association studies (GWASs), greater inclusion of diverse ancestries and low-income countries, the closer integration of psychiatric genetics with electronic medical records, and the development of the neuroscience toolkit for polygenic disorders.
Collapse
Affiliation(s)
- Andrew M McIntosh
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK.
| | - Patrick F Sullivan
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Cathryn M Lewis
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK; Department of Medical and Molecular Genetics, King's College London, London UK
| |
Collapse
|
110
|
Response to the Letter from Dr. Jacob Peedicayil. Psychopharmacology (Berl) 2019; 236:1403-1404. [PMID: 30519767 DOI: 10.1007/s00213-018-5143-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 11/29/2018] [Indexed: 10/27/2022]
|
111
|
Jia P, Chen X, Xie W, Kendler KS, Zhao Z. Mega-analysis of Odds Ratio: A Convergent Method for a Deep Understanding of the Genetic Evidence in Schizophrenia. Schizophr Bull 2019; 45:698-708. [PMID: 29931221 PMCID: PMC6483587 DOI: 10.1093/schbul/sby085] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Numerous high-throughput omics studies have been conducted in schizophrenia, providing an accumulated catalog of susceptible variants and genes. The results from these studies, however, are highly heterogeneous. The variants and genes nominated by different omics studies often have limited overlap with each other. There is thus a pressing need for integrative analysis to unify the different types of data and provide a convergent view of schizophrenia candidate genes (SZgenes). In this study, we collected a comprehensive, multidimensional dataset, including 7819 brain-expressed genes. The data hosted genome-wide association evidence in genetics (eg, genotyping data, copy number variations, de novo mutations), epigenetics, transcriptomics, and literature mining. We developed a method named mega-analysis of odds ratio (MegaOR) to prioritize SZgenes. Application of MegaOR in the multidimensional data resulted in consensus sets of SZgenes (up to 530), each enriched with dense, multidimensional evidence. We proved that these SZgenes had highly tissue-specific expression in brain and nerve and had intensive interactions that were significantly stronger than chance expectation. Furthermore, we found these SZgenes were involved in human brain development by showing strong spatiotemporal expression patterns; these characteristics were replicated in independent brain expression datasets. Finally, we found the SZgenes were enriched in critical functional gene sets involved in neuronal activities, ligand gated ion signaling, and fragile X mental retardation protein targets. In summary, MegaOR analysis reported consensus sets of SZgenes with enriched association evidence to schizophrenia, providing insights into the pathophysiology underlying schizophrenia.
Collapse
Affiliation(s)
- Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX
| | - Xiangning Chen
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV,Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV
| | - Wei Xie
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN
| | - Kenneth S Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA,Department of Psychiatry, Virginia Commonwealth University, Richmond, VA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX,Department of Psychiatry, The University of Texas Health Science Center at Houston, Houston, TX,To whom correspondence should be addressed; School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX 77030, USA; tel: 713-500-3631, fax: 713-500-3907, e-mail:
| |
Collapse
|
112
|
Sullivan PF, Geschwind DH. Defining the Genetic, Genomic, Cellular, and Diagnostic Architectures of Psychiatric Disorders. Cell 2019; 177:162-183. [PMID: 30901538 PMCID: PMC6432948 DOI: 10.1016/j.cell.2019.01.015] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 01/01/2023]
Abstract
Studies of the genetics of psychiatric disorders have become one of the most exciting and fast-moving areas in human genetics. A decade ago, there were few reproducible findings, and now there are hundreds. In this review, we focus on the findings that have illuminated the genetic architecture of psychiatric disorders and the challenges of using these findings to inform our understanding of pathophysiology. The evidence is now overwhelming that psychiatric disorders are "polygenic"-that many genetic loci contribute to risk. With the exception of a subset of those with ASD, few individuals with a psychiatric disorder have a single, deterministic genetic cause; rather, developing a psychiatric disorder is influenced by hundreds of different genetic variants, consistent with a polygenic model. As progressively larger studies have uncovered more about their genetic architecture, the need to elucidate additional architectures has become clear. Even if we were to have complete knowledge of the genetic architecture of a psychiatric disorder, full understanding requires deep knowledge of the functional genomic architecture-the implicated loci impact regulatory processes that influence gene expression and the functional coordination of genes that control biological processes. Following from this is cellular architecture: of all brain regions, cell types, and developmental stages, where and when are the functional architectures operative? Given that the genetic architectures of different psychiatric disorders often strongly overlap, we are challenged to re-evaluate and refine the diagnostic architectures of psychiatric disorders using fundamental genetic and neurobiological data.
Collapse
Affiliation(s)
- Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
| | - Daniel H Geschwind
- Departments of Neurology, Psychiatry, and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Institute for Precision Health, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
113
|
Genomic basis of delayed reward discounting. Behav Processes 2019; 162:157-161. [PMID: 30876880 DOI: 10.1016/j.beproc.2019.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/04/2019] [Accepted: 03/09/2019] [Indexed: 02/07/2023]
Abstract
Delayed reward discounting (DRD) is a behavioral economic measure of impulsivity, reflecting how rapidly a reward loses value based on its temporal distance. In humans, more impulsive DRD is associated with susceptibility to a number of psychiatric diseases (e.g., addiction, ADHD), health outcomes (e.g., obesity), and lifetime outcomes (e.g., educational attainment). Although the determinants of DRD are both genetic and environmental, this review focuses on its genetic basis. Both rodent studies using inbred strains and human twin studies indicate that DRD is moderately heritable, a conclusion that was further supported by a recent human genome-wide association study (GWAS) that used single nucleotide polymorphisms (SNP) to estimate heritability. The GWAS of DRD also identified genetic correlations with psychiatric diagnoses, health outcomes, and measures of cognitive performance. Future research priorities include rodent studies probing putative genetic mechanisms of DRD and human GWASs using larger samples and non-European cohorts. Continuing to characterize genomic influences on DRD has the potential to yield important biological insights with implications for a variety of medically and socially important outcomes.
Collapse
|
114
|
Zhang Z, Cui J, Gao F, Li Y, Zhang G, Liu M, Yan R, Shen Y, Li R. Elevated cleavage of neuregulin-1 by beta-secretase 1 in plasma of schizophrenia patients. Prog Neuropsychopharmacol Biol Psychiatry 2019; 90:161-168. [PMID: 30500411 DOI: 10.1016/j.pnpbp.2018.11.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/23/2018] [Accepted: 11/24/2018] [Indexed: 01/22/2023]
Abstract
Neuregulin 1 (NRG1) is a key candidate susceptibility gene for schizophrenia. It is reported that the function of NRG1 can be regulated by cleavage via the β-Secretase (BACE1), particularly during early development. While current knowledge suggested that schizophrenia might have different phenotypes, it is unknown whether BACE1-cleaved-NRG1 (BACE1-NRG1) activity is related to clinical phenotypes of schizophrenia. In the current study, we used a newly developed enzymatic assay to detect BACE1-NRG1 activity in the human plasma and investigated the levels of cleavage of NRG1 by BACE1 in the plasma from schizophrenia patients. Our results are the first to demonstrate that the level of plasma BACE1-NRG1 activity was significantly increased in subjects affected with schizophrenia compared with healthy controls. Interestingly, the elevated BACE1-NRG1 activity was correlated with the disease severity and duration of schizophrenia, such as patients suffering from shorter-term course and worse disease status expressed higher BACE1-NRG1 activity levels compared to whom with longer duration and less severity of the disease. Furthermore, this is also the first report that the alternation of BACE1-NRG1 activity was a substrate -specific event in schizophrenia. Together, our findings suggested that the plasma BACE1-NRG1 activity can be a potential biomarker for the early diagnosis of schizophrenia.
Collapse
Affiliation(s)
- Zhengrong Zhang
- National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Jie Cui
- Center for Hormone Advanced Science and Education, Roskamp Institute, Sarasota, FL 34243, USA
| | - Feng Gao
- Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Yuhong Li
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Guofu Zhang
- National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Min Liu
- National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Riqiang Yan
- Department of Neurosciences, University of Connecticut School of Medicine, Farmington, CT 06269, USA
| | - Yong Shen
- Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Rena Li
- National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China; Center for Hormone Advanced Science and Education, Roskamp Institute, Sarasota, FL 34243, USA; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
115
|
Sharma S, Ressler KJ. Genomic updates in understanding PTSD. Prog Neuropsychopharmacol Biol Psychiatry 2019; 90:197-203. [PMID: 30452941 PMCID: PMC6431237 DOI: 10.1016/j.pnpbp.2018.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/07/2018] [Accepted: 11/16/2018] [Indexed: 12/14/2022]
Abstract
Twin studies as well as more recent genetics-based heritability analyses demonstrate that up to 40 to 50% of the variance in predicting PTSD following trauma is heritable. However, most of the specific gene pathways and mechanism that mediate risk vs. resilience for PTSD following trauma exposure have yet to be elucidated. This review will examine the latest results from large scale Genome-wide association studies as well as other approaches aimed at understanding mechanisms of development of and recovery from PTSD.
Collapse
Affiliation(s)
- Sumeet Sharma
- Neuroscience Program, Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, United States; McLean Hospital, Division of Depression and Anxiety, Belmont, MA, United States
| | - Kerry J Ressler
- McLean Hospital, Division of Depression and Anxiety, Belmont, MA, United States; Harvard Medical School, Boston, MA, United States; Neuroscience Program, Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, United States.
| |
Collapse
|
116
|
Abstract
After more than 10 years of accumulated efforts, genome-wide association studies (GWAS) have led to many findings, most of which have been deposited into the GWAS Catalog. Between GWAS's inception and March 2017, the GWAS Catalog has collected 2429 studies, 1818 phenotypes, and 28,462 associated SNPs. We reclassified the psychology-related phenotypes into 217 reclassified phenotypes, which accounted for 514 studies and 7052 SNPs. In total, 1223 of the SNPs reached genome-wide significance. Of these, 147 were replicated for the same psychological trait in different studies. Another 305 SNPs were replicated within one original study. The SNPs rs2075650 and rs4420638 were linked to the most replications within a single reclassified phenotype or very similar reclassified phenotypes; both were associated with Alzheimer's disease (AD). Schizophrenia was associated with 74 within-phenotype SNPs reported in independents studies. Alzheimer's disease and schizophrenia were both linked to some physical phenotypes, including cholesterol and body mass index, through common GWAS signals. Alzheimer's disease also shared risk SNPs with age-related phenotypes such as age-related macular degeneration and longevity. Smoking-related SNPs were linked to lung cancer and respiratory function. Alcohol-related SNPs were associated with cardiovascular and digestive system phenotypes and disorders. Two separate studies also identified a shared risk SNP for bipolar disorder and educational attainment. This review revealed a list of reproducible SNPs worthy of future functional investigation. Additionally, by identifying SNPs associated with multiple phenotypes, we illustrated the importance of studying the relationships among phenotypes to resolve the nature of their causal links. The insights within this review will hopefully pave the way for future evidence-based genetic studies.
Collapse
|
117
|
Legge SE, Walters JT. Genetics of clozapine-associated neutropenia: recent advances, challenges and future perspective. Pharmacogenomics 2019; 20:279-290. [PMID: 30767710 PMCID: PMC6563116 DOI: 10.2217/pgs-2018-0188] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Clozapine is the only effective antipsychotic for treatment-resistant schizophrenia but remains widely under prescribed, at least in part due to its potential to cause agranulocytosis and neutropenia. In this article, we provide an overview of the current understanding of the genetics of clozapine-associated agranulocytosis and neutropenia. We now know that the genetic etiology of clozapine-associated neutropenia is complex and is likely to involve variants from several genes including HLA-DQB1, HLA-B and SLCO1B3/SLCO1B7. We describe recent findings relating to the Duffy-null genotype and its association with benign neutropenia in individuals with African ancestry. Further advances will come from sequencing studies, large, cross-population studies and in understanding the molecular mechanisms underlying these associations.
Collapse
Affiliation(s)
- Sophie E Legge
- MRC Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| | - James Tr Walters
- MRC Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| |
Collapse
|
118
|
Wu C, Ding Y, Chen B, Gao Y, Wang Q, Wu Z, Lu L, Luo L, Zhang C, Bao X, Yang P, Fan L, Lei M, Li L. Both Val158Met Polymorphism of Catechol-O-Methyltransferase Gene and Menstrual Cycle Affect Prepulse Inhibition but Not Attentional Modulation of Prepulse Inhibition in Younger-Adult Females. Neuroscience 2019; 404:396-406. [PMID: 30742958 DOI: 10.1016/j.neuroscience.2019.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 12/11/2022]
Abstract
Prepulse inhibition (PPI) can be modulated by both the Val158Met (rs4680) polymorphism of the Catechol-O-Methyltransferase (COMT) gene and the menstrual-cycle-related hormone fluctuations, each of which affects the subcortical/cortical dopamine metabolism. PPI can also be modulated by attention. The attentional modulation of PPI (AMPPI) is sensitive to psychoses. Whether the Val158Met polymorphism affects the AMPPI in female adults at different menstrual-cycle phases is unknown. This study examined whether AMPPI and/or PPI are affected by the Val158Met polymorphism in 177 younger-adult females whose menstrual cycles were mutually different across the menstruation, proliferative, or secretory phases. The AMPPI was evaluated by comparing PPI under the condition of the auditory precedence-effect-induced perceptual spatial separation between the prepulse stimulus and a masking noise (PPIPSS) against that under the condition of the precedence-effect-induced perceptual spatial co-location (PPIPSC). The results showed that both the menstrual cycle and the COMT Val158Met polymorphism affected both PPIPSC and PPIPSS, but not the AMPPI (difference between PPIPSS and PPIPSC). Moreover, throughout the menstrual cycle, both PPIPSC and PPIPSS decreased monotonously in Val/Val-carrier participants. However, the decreasing pattern was not overserved in either Met/Met-carrier or Met/Val-carrier participants. Thus, in healthy younger-adult females, PPIPSC and PPIPSS, but not the AMPPI, is vulnerable to changes of ovarian hormones, and the COMT Val158Met polymorphism also has a modulating effect on this menstrual-cycle-dependent PPI variation. In contrast, the AMPPI seems to be more steadily trait-based, less vulnerable to ovarian hormone fluctuations, and may be useful in assisting the diagnosis of schizophrenia in female adults.
Collapse
Affiliation(s)
- Chao Wu
- School of Nursing, Peking University Health Science Center, Beijing, 100191, China; PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Yu Ding
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Biqing Chen
- PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Yayue Gao
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Qian Wang
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Zhemeng Wu
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Lingxi Lu
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Lu Luo
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Changxin Zhang
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Xiaohan Bao
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Pengcheng Yang
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Langchen Fan
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Ming Lei
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China
| | - Liang Li
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Speech and Hearing Research Center, Key Laboratory on Machine Perception (Ministry of Education), Peking University, Beijing 100080, China; Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
119
|
Imputation of behavioral candidate gene repeat variants in 486,551 publicly-available UK Biobank individuals. Eur J Hum Genet 2019; 27:963-969. [PMID: 30723318 DOI: 10.1038/s41431-019-0349-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 01/09/2019] [Accepted: 01/16/2019] [Indexed: 02/06/2023] Open
Abstract
Some of the most widely studied variants in psychiatric genetics include variable number tandem repeat variants (VNTRs) in SLC6A3, DRD4, SLC6A4, and MAOA. While initial findings suggested large effects, their importance with respect to psychiatric phenotypes is the subject of much debate with broadly conflicting results. Despite broad interest, these loci remain absent from the largest available samples, such as the UK Biobank, limiting researchers' ability to test these contentious hypotheses rigorously in large samples. Here, using two independent reference datasets, we report out-of-sample imputation accuracy estimates of >0.96 for all four VNTR variants and one modifying SNP, depending on the reference and target dataset. We describe the imputation procedures of these candidate variants in 486,551 UK Biobank individuals, and have made the imputed variant data available to UK Biobank researchers. This resource, provided to the scientific community, will allow the most rigorous tests to-date of the roles of these variants in behavioral and psychiatric phenotypes.
Collapse
|
120
|
Rhoades R, Jackson F, Teng S. Discovery of rare variants implicated in schizophrenia using next-generation sequencing. JOURNAL OF TRANSLATIONAL GENETICS AND GENOMICS 2019; 3:1-20. [PMID: 33981965 PMCID: PMC8112455 DOI: 10.20517/jtgg.2018.26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Schizophrenia is a highly heritable psychiatric disorder that affects 1% of the population. Genome-wide association studies have identified common variants in candidate genes associated with schizophrenia, but the genetics mechanisms of this disorder have not yet been elucidated. The discovery of rare genetic variants that contribute to schizophrenia symptoms promises to help explain the missing heritability of the disease. Next generation sequencing techniques are revolutionizing the field of psychiatric genetics. Various statistical approaches have been developed for rare variant association testing in case-control and family studies. Targeted resequencing, whole exome sequencing and whole genome sequencing combined with these computational tools are used for the discovery of rare genetic variations in schizophrenia. The findings provide useful information for characterizing the rare mutations and elucidating the genetic mechanisms by which the variants cause schizophrenia.
Collapse
Affiliation(s)
- Raina Rhoades
- Department of Biology, Howard University, Washington, DC 20059, USA
| | - Fatimah Jackson
- Department of Biology, Howard University, Washington, DC 20059, USA
| | - Shaolei Teng
- Department of Biology, Howard University, Washington, DC 20059, USA
| |
Collapse
|
121
|
Winship IR, Dursun SM, Baker GB, Balista PA, Kandratavicius L, Maia-de-Oliveira JP, Hallak J, Howland JG. An Overview of Animal Models Related to Schizophrenia. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2019; 64:5-17. [PMID: 29742910 PMCID: PMC6364139 DOI: 10.1177/0706743718773728] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Schizophrenia is a heterogeneous psychiatric disorder that is poorly treated with current therapies. In this brief review, we provide an update regarding the use of animal models to study schizophrenia in an attempt to understand its aetiology and develop novel therapeutic strategies. Tremendous progress has been made developing and validating rodent models that replicate the aetiologies, brain pathologies, and behavioural abnormalities associated with schizophrenia in humans. Here, models are grouped into 3 categories-developmental, drug induced, and genetic-to reflect the heterogeneous risk factors associated with schizophrenia. Each of these models is associated with varied but overlapping pathophysiology, endophenotypes, behavioural abnormalities, and cognitive impairments. Studying schizophrenia using multiple models will permit an understanding of the core features of the disease, thereby facilitating preclinical research aimed at the development and validation of better pharmacotherapies to alter the progression of schizophrenia or alleviate its debilitating symptoms.
Collapse
Affiliation(s)
- Ian R Winship
- 1 Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta
| | - Serdar M Dursun
- 2 Department of Psychiatry, Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta.,3 National Institute of Science and Technology-Translational Science, Brazil
| | - Glen B Baker
- 2 Department of Psychiatry, Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta.,3 National Institute of Science and Technology-Translational Science, Brazil
| | - Priscila A Balista
- 4 Department of Pharmacy, Centro Universitario das Faculdades Metropolitanas Unidas, São Paulo, Brazil
| | - Ludmyla Kandratavicius
- 5 Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Joao Paulo Maia-de-Oliveira
- 3 National Institute of Science and Technology-Translational Science, Brazil.,6 Department of Clinical Medicine, Rio Grande do Norte Federal University, Natal, Brazil
| | - Jaime Hallak
- 3 National Institute of Science and Technology-Translational Science, Brazil.,5 Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil.,7 Department of Psychiatry (NRU), University of Alberta, Edmonton, Alberta
| | - John G Howland
- 8 Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan
| |
Collapse
|
122
|
Gianfrancesco O, Bubb VJ, Quinn JP. Treating the "E" in "G × E": Trauma-Informed Approaches and Psychological Therapy Interventions in Psychosis. Front Psychiatry 2019; 10:9. [PMID: 30761022 PMCID: PMC6363686 DOI: 10.3389/fpsyt.2019.00009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/08/2019] [Indexed: 12/31/2022] Open
Abstract
Despite advances in genetic research, causal variants affecting risk for schizophrenia remain poorly characterized, and the top 108 loci identified through genome-wide association studies (GWAS) explain only 3.4% of variance in risk profiles. Such work is defining the highly complex nature of this condition, with omnigenic models of schizophrenia suggesting that gene regulatory networks are sufficiently interconnected such that altered expression of any "peripheral" gene in a relevant cell type has the capacity to indirectly modulate the expression of "core" schizophrenia-associated genes. This wealth of associated genes with small effect sizes makes identifying new druggable targets difficult, and current pharmacological treatments for schizophrenia can involve serious side effects. However, the fact that the majority of schizophrenia genome-wide associated variants fall within non-coding DNA is suggestive of their potential to modulate gene regulation. This would be consistent with risks that can be mediated in a "gene × environment" (G × E) manner. Stress and trauma can alter the regulation of key brain-related pathways over the lifetime of an individual, including modulation of brain development, and neurochemistry in the adult. Recent studies demonstrate a significant overlap between psychotic symptoms and trauma, ranging from prior trauma contributing to psychosis, as well as trauma in response to the experience of psychosis itself or in response to treatment. Given the known effects of trauma on both CNS gene expression and severity of psychosis symptoms, it may be that pharmacological treatment alone risks leaving individuals with a highly stressful and unresolved environmental component that continues to act in a "G × E" manner, with the likelihood that this would negatively impact recovery and relapse risk. This review aims to cover the recent advances elucidating the complex genetic architecture of schizophrenia, as well as the long-term effects of early life trauma on brain function and future mental health risk. Further, the evidence demonstrating the role of ongoing responses to trauma or heightened stress sensitivity, and their impact on the course of illness and recovery, is presented. Finally, the need for trauma-informed approaches and psychological therapy-based interventions is discussed, and a brief overview of the evidence to determine their utility is presented.
Collapse
Affiliation(s)
- Olympia Gianfrancesco
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom.,MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Vivien J Bubb
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - John P Quinn
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
123
|
Meng Q, Wang K, Brunetti T, Xia Y, Jiao C, Dai R, Fitzgerald D, Thomas A, Jay L, Eckart H, Grennan K, Imamura-Kawasawa Y, Li M, Sestan N, White KP, Chen C, Liu C. The DGCR5 long noncoding RNA may regulate expression of several schizophrenia-related genes. Sci Transl Med 2018; 10:eaat6912. [PMID: 30545965 PMCID: PMC6487854 DOI: 10.1126/scitranslmed.aat6912] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 10/22/2018] [Indexed: 12/12/2022]
Abstract
A number of studies indicate that rare copy number variations (CNVs) contribute to the risk of schizophrenia (SCZ). Most of these studies have focused on protein-coding genes residing in the CNVs. Here, we investigated long noncoding RNAs (lncRNAs) within 10 SCZ risk-associated CNV deletion regions (CNV-lncRNAs) and examined their potential contribution to SCZ risk. We used RNA sequencing transcriptome data derived from postmortem brain tissue from control individuals without psychiatric disease as part of the PsychENCODE BrainGVEX and Developmental Capstone projects. We carried out weighted gene coexpression network analysis to identify protein-coding genes coexpressed with CNV-lncRNAs in the human brain. We identified one neuronal function-related coexpression module shared by both datasets. This module contained a lncRNA called DGCR5 within the 22q11.2 CNV region, which was identified as a hub gene. Protein-coding genes associated with SCZ genome-wide association study signals, de novo mutations, or differential expression were also contained in this neuronal module. Using DGCR5 knockdown and overexpression experiments in human neural progenitor cells derived from human induced pluripotent stem cells, we identified a potential role for DGCR5 in regulating certain SCZ-related genes.
Collapse
Affiliation(s)
- Qingtuan Meng
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Kangli Wang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Tonya Brunetti
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yan Xia
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Chuan Jiao
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Rujia Dai
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Dominic Fitzgerald
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Amber Thomas
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Lindsey Jay
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Heather Eckart
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Kay Grennan
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yuka Imamura-Kawasawa
- Departments of Pharmacology and Biochemistry and Molecular Biology, Institute for Personalized Medicine, Penn State College of Medicine, Hershey, PA 17033, USA
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Mingfeng Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Nenad Sestan
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Kevin P White
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
- Tempus Labs Inc., Chicago, IL 60654, USA
| | - Chao Chen
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chunyu Liu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
- School of Psychology, Shaanxi Normal University, Xian, Shaanxi, China
| |
Collapse
|
124
|
Lucatch AM, Lowe DJE, Clark RC, Kozak K, George TP. Neurobiological Determinants of Tobacco Smoking in Schizophrenia. Front Psychiatry 2018; 9:672. [PMID: 30574101 PMCID: PMC6291492 DOI: 10.3389/fpsyt.2018.00672] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/21/2018] [Indexed: 12/22/2022] Open
Abstract
Purpose of review: To provide an overview of the underlying neurobiology of tobacco smoking in schizophrenia, and implications for treatment of this comorbidity. Recent findings: Explanations for heavy tobacco smoking in schizophrenia include pro-cognitive effects of nicotine, and remediation of the underlying pathophysiology of schizophrenia. Nicotine may ameliorate neurochemical deficits through nicotine acetylcholine receptors (nAChRs) located on the dopamine, glutamate, and GABA neurons. Neurophysiological indices including electroencephalography, electromyography, and smooth pursuit eye movement (SPEM) paradigms may be biomarkers for underlying neuronal imbalances that contribute to the specific risk of tobacco smoking initiation, maintenance, and difficulty quitting within schizophrenia. Moreover, several social factors including socioeconomic factors and permissive smoking culture in mental health facilities, may contribute to the smoking behaviors (initiation, maintenance, and inability to quit smoking) within this disorder. Summary: Tobacco smoking may alleviate specific symptoms associated with schizophrenia. Understanding the neurobiological underpinnings and psychosocial determinants of this comorbidity may better explain these potential beneficial effects, while also providing important insights into effective treatments for smoking cessation.
Collapse
Affiliation(s)
- Aliya M. Lucatch
- Addictions Division, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Darby J. E. Lowe
- Addictions Division, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Rachel C. Clark
- Addictions Division, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Karolina Kozak
- Addictions Division, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Tony P. George
- Addictions Division, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Division and Brain and Therapeutics, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
125
|
Kannan G, Prandovszky E, Severance E, Yolken RH, Pletnikov MV. A New T. gondii Mouse Model of Gene-Environment Interaction Relevant to Psychiatric Disease. SCIENTIFICA 2018; 2018:7590958. [PMID: 30631636 PMCID: PMC6305013 DOI: 10.1155/2018/7590958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/11/2018] [Indexed: 06/09/2023]
Abstract
Infection with the protozoan parasite, Toxoplasma gondii (T. gondii), was linked to several psychiatric disorders. The exact mechanisms of a hypothesized contribution of T. gondii infection are poorly understood, and it appears that only a subset of seropositive individuals go on to develop a mental illness, suggesting genetic vulnerability. In order to stimulate mechanistic studies of how exposure to T. gondii could interact with genetic predisposition to psychiatric disorders, we have generated and characterized a mouse model of chronic T. gondii infection in BALB/c mice with inducible forebrain neuronal expression of a C-terminus truncated dominant-negative form of disrupted-in-schizophrenia 1 (DN-DISC1). In this gene-environment interaction (GxE) model, exposing control and DN-DISC1 male and female mice to T. gondii produced sex-dependent abnormalities in locomotor activity and prepulse inhibition of the acoustic startle. No genotype- or sex-dependent effects were found on levels of anti-Toxoplasma IgG antibodies or anti-NMDAR or C1q antibodies. Our work demonstrates that a psychiatric genetic risk factor, DN-DISC1, modulates the neurobehavioral effects of chronic T. gondii infection in a sex-dependent manner. The present T. gondii model of GxE provides a valuable experimental system for future mechanistic studies and evaluation of new treatments.
Collapse
Affiliation(s)
- Geetha Kannan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Emese Prandovszky
- Stanley Neurovirology Laboratory, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Emily Severance
- Stanley Neurovirology Laboratory, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert H. Yolken
- Stanley Neurovirology Laboratory, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mikhail V. Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
126
|
Oishi K, Kanahara N, Takase M, Oda Y, Nakata Y, Niitsu T, Ishikawa M, Sato Y, Iyo M. Vulnerable combinations of functional dopaminergic polymorphisms to late-onset treatment resistant schizophrenia. PLoS One 2018; 13:e0207133. [PMID: 30408108 PMCID: PMC6224074 DOI: 10.1371/journal.pone.0207133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/25/2018] [Indexed: 11/19/2022] Open
Abstract
Background A significant portion of patients with schizophrenia who respond to initial antipsychotic treatment acquire treatment resistance. One of the possible pathogeneses of treatment-resistant schizophrenia (TRS) is antipsychotic-induced dopamine supersensitivity psychosis (Ai-DSP). Patients with this disease progression might share some genetic vulnerabilities, and thus determining individuals with higher risks of developing Ai-DSP could contribute to preventing iatrogenic development of TRS. Therefore, we decided to examine whether combinations of functional single nucleotide polymorphisms (SNPs) known to affect dopaminergic functions are related to Ai-DSP development. Methods In this case-control study, 357 Japanese participants diagnosed with schizophrenia or schizoaffective disorder were recruited and divided into two groups, those with and without Ai-DSP. As functional SNPs, we examined rs10770141 of the tyrosine hydroxylase gene, rs4680 of the catechol-O-methyltransferase gene, and rs1799732 and rs1800497 of the DRD2 genes, which are known to possess strong directional ties to dopamine synthesis, dopamine degradation and post-synaptic DRD2 prevalence, respectively. Results Among the 357 Japanese patients with schizophrenia or schizoaffective disorder, 130 were classified as Ai-DSP(+) and the other 227 as Ai-DSP(-). Significantly higher proportions of Ai-DSP(+) patients were found to have the SNP combinations of rs10770141/rs4680 (57.9%, OR2.654, 95%CI1.036–6.787, P = 0.048) and rs10770141/rs4680/ rs1800497 (64.3%, OR4.230, 95%CI1.306–13.619, P = 0.029). However, no single SNP was associated with Ai-DSP. Conclusions We preliminarily found that carrying particular combinations of functional SNPs, which are related to relatively higher dopamine synthesis and dopamine degradation and lower naïve DRD2, might indicate vulnerability to development of Ai-DSP. However, further studies are needed to validate the present results.
Collapse
Affiliation(s)
- Kengo Oishi
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chuou-ku, Chiba, Chiba, Japan
- * E-mail:
| | - Nobuhisa Kanahara
- Division of Medical Treatment and Rehabilitation, Chiba University Center for Forensic Mental Health, Chuou-ku, Chiba, Chiba, Japan
| | - Masayuki Takase
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chuou-ku, Chiba, Chiba, Japan
- Zucker Hillside Hospital, Glen Oaks, NY, United States of America
| | - Yasunori Oda
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chuou-ku, Chiba, Chiba, Japan
| | - Yusuke Nakata
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chuou-ku, Chiba, Chiba, Japan
| | - Tomihisa Niitsu
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chuou-ku, Chiba, Chiba, Japan
| | - Masatomo Ishikawa
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chuou-ku, Chiba, Chiba, Japan
| | - Yasunori Sato
- Department of Global Clinical Research, Chiba University Graduate School of Medicine, Chuou-ku, Chiba, Chiba, Japan
| | - Masaomi Iyo
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chuou-ku, Chiba, Chiba, Japan
| |
Collapse
|
127
|
Altered hippocampal gene expression and structure in transgenic mice overexpressing neuregulin 1 (Nrg1) type I. Transl Psychiatry 2018; 8:229. [PMID: 30348978 PMCID: PMC6197224 DOI: 10.1038/s41398-018-0288-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/24/2018] [Accepted: 09/26/2018] [Indexed: 11/26/2022] Open
Abstract
Transgenic mice overexpressing the type I isoform of neuregulin 1 (Nrg1; NRG1) have alterations in hippocampal gamma oscillations and an age-emergent deficit in hippocampus-dependent spatial working memory. Here, we examined the molecular and morphological correlates of these findings. Microarrays showed over 100 hippocampal transcripts differentially expressed in Nrg1tg-type I mice, with enrichment of genes related to neuromodulation and, in older mice, of genes involved in inflammation and immunity. Nrg1tg-type I mice had an enlarged hippocampus with a widened dentate gyrus. The results show that Nrg1 type I impacts on hippocampal gene expression and structure in a multifaceted and partly age-related way, complementing the evidence implicating Nrg1 signaling in aspects of hippocampal function. The findings are also relevant to the possible role of NRG1 signaling in the pathophysiology of schizophrenia or other disorders affecting this brain region.
Collapse
|
128
|
Dahoun T, Pardiñas AF, Veronese M, Bloomfield MAP, Jauhar S, Bonoldi I, Froudist-Walsh S, Nosarti C, Korth C, Hennah W, Walters J, Prata D, Howes OD. The effect of the DISC1 Ser704Cys polymorphism on striatal dopamine synthesis capacity: an [18F]-DOPA PET study. Hum Mol Genet 2018; 27:3498-3506. [PMID: 29945223 PMCID: PMC6168972 DOI: 10.1093/hmg/ddy242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/22/2018] [Accepted: 06/22/2018] [Indexed: 11/14/2022] Open
Abstract
Whilst the role of the Disrupted-in-Schizophrenia 1 (DISC1) gene in the aetiology of major mental illnesses is debated, the characterization of its function lends it credibility as a candidate. A key aspect of this functional characterization is the determination of the role of common non-synonymous polymorphisms on normal variation within these functions. The common allele (A) of the DISC1 single-nucleotide polymorphism (SNP) rs821616 encodes a serine (ser) at the Ser704Cys polymorphism, and has been shown to increase the phosphorylation of extracellular signal-regulated protein Kinases 1 and 2 (ERK1/2) that stimulate the phosphorylation of tyrosine hydroxylase, the rate-limiting enzyme for dopamine biosynthesis. We therefore set out to test the hypothesis that human ser (A) homozygotes would show elevated dopamine synthesis capacity compared with cysteine (cys) homozygotes and heterozygotes (TT and AT) for rs821616. [18F]-DOPA positron emission tomography (PET) was used to index striatal dopamine synthesis capacity as the influx rate constant Kicer in healthy volunteers DISC1 rs821616 ser homozygotes (N = 46) and healthy volunteers DISC1 rs821616 cys homozygotes and heterozygotes (N = 56), matched for age, gender, ethnicity and using three scanners. We found DISC1 rs821616 ser homozygotes exhibited a significantly higher striatal Kicer compared with cys homozygotes and heterozygotes (P = 0.012) explaining 6.4% of the variance (partial η2 = 0.064). Our finding is consistent with its previous association with heightened activation of ERK1/2, which stimulates tyrosine hydroxylase activity for dopamine synthesis. This could be a potential mechanism mediating risk for psychosis, lending further credibility to the fact that DISC1 is of functional interest in the aetiology of major mental illness.
Collapse
Affiliation(s)
- Tarik Dahoun
- Psychiatric Imaging Group, Robert Steiner MRI Unit, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
- Faculty of Medicine, Institute of Clinical Sciences (ICS), Imperial College London, Hammersmith Hospital, London, UK
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford OX37 JX, UK
| | - Antonio F Pardiñas
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, King’s College London, London, UK
| | - Michael A P Bloomfield
- Psychiatric Imaging Group, Robert Steiner MRI Unit, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
- Faculty of Medicine, Institute of Clinical Sciences (ICS), Imperial College London, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King’s College London, London, UK
- Division of Psychiatry, University College London, London, UK
- Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Sameer Jauhar
- Psychiatric Imaging Group, Robert Steiner MRI Unit, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
- Faculty of Medicine, Institute of Clinical Sciences (ICS), Imperial College London, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King’s College London, London, UK
| | - Ilaria Bonoldi
- Psychiatric Imaging Group, Robert Steiner MRI Unit, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
- Faculty of Medicine, Institute of Clinical Sciences (ICS), Imperial College London, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King’s College London, London, UK
| | | | - Chiara Nosarti
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King’s College London, London, UK
- Division of Imaging Sciences & Biomedical Engineering, Centre for the Developing Brain, King’s College London, London, UK
| | - Carsten Korth
- Department Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - William Hennah
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
- Mental Health Unit, Department of Health, National Institute for Health and Welfare, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
| | - James Walters
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Diana Prata
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Instituto Universitário de Lisboa (ISCTE-IUL), Cis-IUL, Lisbon, Portugal
| | - Oliver D Howes
- Psychiatric Imaging Group, Robert Steiner MRI Unit, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
- Faculty of Medicine, Institute of Clinical Sciences (ICS), Imperial College London, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King’s College London, London, UK
| |
Collapse
|
129
|
Guo X, Yang J, Huang J, Chen Z, Wu X, Zhu L, Huang G, Long J, Su L. Influence of CTNNB1 rs2953 polymorphism on schizophrenia susceptibility in Chinese Han population through modifying miR-485 binding to CTNNB1. GENES BRAIN AND BEHAVIOR 2018; 18:e12524. [PMID: 30280518 DOI: 10.1111/gbb.12524] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/28/2018] [Indexed: 12/21/2022]
Abstract
Schizophrenia (SCZ) and bipolar disorder (BD) are two major neuropsychiatric diseases that are the most substantial causes of disability and mortality worldwide. CTNNB1 encodes beta-catenin, an important protein in canonical Wnt signaling. We aimed to investigate the association between the rs2953 of CTNNB1 and the risk of SCZ and BD and to further explore the function of rs2953. A total of 1658 samples (548 SCZ cases, 512 BD cases, and 598 controls) were examined in terms of the genotype of CTNNB1 rs2953. The mRNA expression level of CTNNB1 significantly increased in the SCZ and BD groups compared with that in the control group. Significant association remained between CTNNB1 3'-untranslated region (UTR) variant rs2953 and SCZ susceptibility (additive and dominant model) after gender and age were adjusted. rs2953 disrupted the binding of CTNNB1 and miR-485. miR-485 significantly suppressed the luciferase activity of CTNNB1-T vector by binding to the CTNNB1 3'-UTR containing the T allele of rs2953. The mRNA expression of CTNNB1 can be used as a biomarker for the diagnosis of SCZ and BD. The 3'-UTR variant rs2953 in CTNNB1 influences the risk of SCZ in the Han Chinese population and modifies the binding of miR-485 to CTNNB1.
Collapse
Affiliation(s)
- Xiaojing Guo
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| | - Jialei Yang
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiao Huang
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhaoxia Chen
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| | - Xulong Wu
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| | - Lulu Zhu
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| | - Guifeng Huang
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| | - Jianxiong Long
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| | - Li Su
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
130
|
Abstract
Schizophrenia and other types of psychosis incur suffering, high health care costs and loss of human potential, due to the combination of early onset and poor response to treatment. Our ability to prevent or cure psychosis depends on knowledge of causal mechanisms. Molecular genetic studies show that thousands of common and rare variants contribute to the genetic risk for psychosis. Epidemiological studies have identified many environmental factors associated with increased risk of psychosis. However, no single genetic or environmental factor is sufficient to cause psychosis on its own. The risk of developing psychosis increases with the accumulation of many genetic risk variants and exposures to multiple adverse environmental factors. Additionally, the impact of environmental exposures likely depends on genetic factors, through gene-environment interactions. Only a few specific gene-environment combinations that lead to increased risk of psychosis have been identified to date. An example of replicable gene-environment interaction is a common polymorphism in the AKT1 gene that makes its carriers sensitive to developing psychosis with regular cannabis use. A synthesis of results from twin studies, molecular genetics, and epidemiological research outlines the many genetic and environmental factors contributing to psychosis. The interplay between these factors needs to be considered to draw a complete picture of etiology. To reach a more complete explanation of psychosis that can inform preventive strategies, future research should focus on longitudinal assessments of multiple environmental exposures within large, genotyped cohorts beginning early in life.
Collapse
Affiliation(s)
- Alyson Zwicker
- Department of Pathology,Dalhousie University,Halifax,NS,Canada
| | | | - Rudolf Uher
- Department of Pathology,Dalhousie University,Halifax,NS,Canada
| |
Collapse
|
131
|
Arthur VL, Shuldiner E, Remmers EF, Hinks A, Grom AA, Foell D, Martini A, Gattorno M, Özen S, Prahalad S, Zeft AS, Bohnsack JF, Ilowite NT, Mellins ED, Russo R, Len C, Oliveira S, Yeung RSM, Rosenberg AM, Wedderburn LR, Anton J, Haas JP, Rösen-Wolff A, Minden K, Szymanski AM, Thomson W, Kastner DL, Woo P, Ombrello MJ. IL1RN Variation Influences Both Disease Susceptibility and Response to Recombinant Human Interleukin-1 Receptor Antagonist Therapy in Systemic Juvenile Idiopathic Arthritis. Arthritis Rheumatol 2018; 70:1319-1330. [PMID: 29609200 PMCID: PMC6105455 DOI: 10.1002/art.40498] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/13/2018] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To determine whether systemic juvenile idiopathic arthritis (JIA) susceptibility loci that were identified by candidate gene studies demonstrate association with systemic JIA in the largest study population assembled to date. METHODS Single-nucleotide polymorphisms (SNPs) from 11 previously reported systemic JIA risk loci were examined for association in 9 populations, including 770 patients with systemic JIA and 6,947 controls. The effect of systemic JIA-associated SNPs on gene expression was evaluated in silico in paired whole genome and RNA sequencing data from the lymphoblastoid cell lines (LCLs) of 373 European subjects from the 1000 Genomes Project. Responses of systemic JIA-associated SNPs to anakinra treatment were evaluated in 38 US patients for whom treatment response data were available. RESULTS We found no association between the previously reported 26 SNPs and systemic JIA. Expanded analysis of the regions containing the 26 SNPs revealed only 1 significant association: the promoter region of IL1RN (P < 1 × 10-4 ). Systemic JIA-associated SNPs correlated with IL1RN expression in LCLs, with an inverse correlation between systemic JIA risk and IL1RN expression. The presence of homozygous IL1RN high expression alleles correlated strongly with a lack of response to anakinra therapy (odds ratio 28.7 [95% confidence interval 3.2-255.8]). CONCLUSION In our study, IL1RN was the only candidate locus associated with systemic JIA. The implicated SNPs are among the strongest known determinants of IL1RN and interleukin-1 receptor antagonist levels, linking low expression with increased systemic JIA risk. Homozygous high expression alleles predicted nonresponsiveness to anakinra therapy, making them ideal candidate biomarkers to guide systemic JIA treatment. This study is an important first step toward the personalized treatment of systemic JIA.
Collapse
Affiliation(s)
- Victoria L. Arthur
- Translational Genetics and Genomics Unit, National Institute of
Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, US
Department of Health & Human Services, Bethesda, MD, USA
| | - Emily Shuldiner
- Translational Genetics and Genomics Unit, National Institute of
Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, US
Department of Health & Human Services, Bethesda, MD, USA
| | - Elaine F. Remmers
- Inflammatory Disease Section, National Human Genome Research
Institute, National Institutes of Health, US Department of Health & Human
Services, Bethesda, MD, USA
| | - Anne Hinks
- Arthritis Research UK Centre for Genetics and Genomics, Centre for
Musculoskeletal Research, University of Manchester, Manchester, UK
| | - Alexei A. Grom
- Department of Pediatrics, University of Cincinnati, College of
Medicine, Cincinnati, OH, USA
- Cincinnati Children’s Hospital Medical Center, Cincinnati,
OH, USA
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University
Hospital Münster, Münster, Germany
| | - Alberto Martini
- Direzione Scientifica, G. Gaslini Institute, Genoa, Italy
- Clinica Pediatrica e Reumatologia, G. Gaslini Institute and
University of Genoa, Genoa, Italy
| | - Marco Gattorno
- Clinica Pediatrica e Reumatologia, G. Gaslini Institute and
University of Genoa, Genoa, Italy
| | - Seza Özen
- Department of Pediatric Rheumatology, Hacettepe University, Ankara,
Turkey
| | - Sampath Prahalad
- Departments of Pediatrics and Human Genetics, Emory University
School of Medicine, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA
| | - Andrew S. Zeft
- Department of Pediatrics, Cleveland Clinic, Cleveland, OH,
USA
| | - John F. Bohnsack
- Department of Pediatrics, University of Utah, Salt Lake City, UT,
USA
| | - Norman T. Ilowite
- Department of Pediatrics, Albert Einstein College of Medicine and
Children’s Hospital at Montefiore, Bronx, NY, USA
| | | | - Ricardo Russo
- Service of Immunology and Rheumatology, Hospital de Pediatria
Garrahan, Buenos Aires, Argentina
| | - Claudio Len
- Department of Pediatrics, Universidade Federal de São
Paulo, São Paulo, Brazil
| | - Sheila Oliveira
- Universidade Federal de Rio de Janeiro, Rio de Janeiro,
Brazil
| | - Rae S. M. Yeung
- Department of Pediatrics, University of Toronto, Toronto,
Canada
- Department of Immunology, University of Toronto, Toronto,
Canada
- Institute of Medical Science, University of Toronto, Toronto,
Canada
| | - Alan M. Rosenberg
- Department of Pediatrics, University of Saskatchewan, Saskatoon,
Canada
| | - Lucy R. Wedderburn
- University College London Great Ormond Street Hospital Institute of
Child Health, University College London, London, UK
- Center of Paediatric and Adolescent Rheumatology, University
College London, London, UK
- NIHR GOSH Biomedical Research Centre, London, UK
| | - Jordi Anton
- Pediatric Rheumatology Unit, Hospital Sant Joan de Déu,
Universitat de Barcelona, Barcelona, Spain
| | - Johannes-Peter Haas
- German Center for Pediatric and Adolescent Rheumatology,
Garmisch-Partenkirchen, Germany
| | | | - Kirsten Minden
- Charité University Medicine, Berlin, Germany
- German Rheumatism Research Centre, Epidemiology Unit, Berlin,
Germany
| | - Ann Marie Szymanski
- Translational Genetics and Genomics Unit, National Institute of
Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, US
Department of Health & Human Services, Bethesda, MD, USA
| | | | - Wendy Thomson
- Arthritis Research UK Centre for Genetics and Genomics, Centre for
Musculoskeletal Research, University of Manchester, Manchester, UK
- National Institute for Health Research Manchester Biomedical
Centre, Central Manchester National Health Service Foundation Trust, Manchester
Academic Health Centre, University of Manchester, Manchester, UK
| | - Daniel L. Kastner
- Inflammatory Disease Section, National Human Genome Research
Institute, National Institutes of Health, US Department of Health & Human
Services, Bethesda, MD, USA
| | - Patricia Woo
- University College London Great Ormond Street Hospital Institute of
Child Health, University College London, London, UK
| | - Michael J. Ombrello
- Translational Genetics and Genomics Unit, National Institute of
Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, US
Department of Health & Human Services, Bethesda, MD, USA
| |
Collapse
|
132
|
Gurriarán X, Rodríguez-López J, Flórez G, Pereiro C, Fernández JM, Fariñas E, Estévez V, Arrojo M, Costas J. Relationships between substance abuse/dependence and psychiatric disorders based on polygenic scores. GENES BRAIN AND BEHAVIOR 2018; 18:e12504. [PMID: 29974660 DOI: 10.1111/gbb.12504] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/20/2018] [Accepted: 07/02/2018] [Indexed: 12/25/2022]
Abstract
Genetic susceptibility to substance use disorders (SUDs) is partially shared between substances. Heritability of any substance dependence, estimated as 54%, is partly explained by additive effects of common variants. Comorbidity between SUDs and other psychiatric disorders is frequent. The present study aims to analyze the additive role of common variants in this comorbidity using polygenic scores (PGSs) based on genome-wide association study discovery samples of schizophrenia (SCZ), bipolar disorder, attention-deficit/hyperactivity disorder, autism spectrum disorder, major depressive disorder and anxiety disorders, available from large consortia. PGSs were calculated for 534 patients meeting DSM-IV criteria for dependence of a substance and abuse/dependence of another substance between alcohol, tobacco, cannabis, cocaine, opiates, hypnotics, stimulants, hallucinogens and solvents; and 587 blood donors from the same population, Iberians from Galicia, as controls. Significance of the PGS and percentage of variance explained were calculated by logistic regression. Using discovery samples of similar size, significant associations with SUDs were detected for SCZ PGS. SCZ PGS explained more variance in SUDs than in most psychiatric disorders. Cross-disorder PGS based on five psychiatric disorders was significant after adjustment for the effect of SCZ PGS. SCZ PGS was significantly higher in women than in men abusing alcohol. Our findings indicate that SUDs share genetic susceptibility with SCZ to a greater extent than with other psychiatric disorders, including externalizing disorders such as attention-deficit/hyperactivity disorder. Women have lower probability to develop substance abuse/dependence than men at similar PGS probably because of a higher social pressure against excessive drug use in women.
Collapse
Affiliation(s)
- Xaquín Gurriarán
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain
| | - Julio Rodríguez-López
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain
| | - Gerardo Flórez
- Unidade de Conductas Adictivas, Servizo de Psiquiatría, Complexo Hospitalario Universitario de Ourense (CHUO), Servizo Galego de Saúde (SERGAS), Ourense, Galicia, Spain
| | - César Pereiro
- Unidade Asistencial de Drogodependencias (ACLAD), A Coruña, Galicia, Spain
| | - José M Fernández
- Unidade Asistencial de Drogodependencias, Ribeira, Galicia, Spain
| | - Emilio Fariñas
- Unidade Municipal de Atención a Drogodependientes (UMAD), Santiago de Compostela, Galicia, Spain
| | - Valentín Estévez
- Unidade de Conductas Adictivas, Servizo de Psiquiatría, Complexo Hospitalario Universitario de Ourense (CHUO), Servizo Galego de Saúde (SERGAS), Ourense, Galicia, Spain
| | - Manuel Arrojo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain.,Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain
| | - Javier Costas
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain
| | | |
Collapse
|
133
|
Keller MC. Evolutionary Perspectives on Genetic and Environmental Risk Factors for Psychiatric Disorders. Annu Rev Clin Psychol 2018; 14:471-493. [DOI: 10.1146/annurev-clinpsy-050817-084854] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Matthew C. Keller
- Department of Psychology and Neuroscience and the Institute for Behavioral Genetics, University of Colorado at Boulder, Boulder, Colorado 80309, USA
| |
Collapse
|
134
|
An analytical framework for whole-genome sequence association studies and its implications for autism spectrum disorder. Nat Genet 2018; 50:727-736. [PMID: 29700473 DOI: 10.1038/s41588-018-0107-y] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 03/06/2018] [Indexed: 11/08/2022]
Abstract
Genomic association studies of common or rare protein-coding variation have established robust statistical approaches to account for multiple testing. Here we present a comparable framework to evaluate rare and de novo noncoding single-nucleotide variants, insertion/deletions, and all classes of structural variation from whole-genome sequencing (WGS). Integrating genomic annotations at the level of nucleotides, genes, and regulatory regions, we define 51,801 annotation categories. Analyses of 519 autism spectrum disorder families did not identify association with any categories after correction for 4,123 effective tests. Without appropriate correction, biologically plausible associations are observed in both cases and controls. Despite excluding previously identified gene-disrupting mutations, coding regions still exhibited the strongest associations. Thus, in autism, the contribution of de novo noncoding variation is probably modest in comparison to that of de novo coding variants. Robust results from future WGS studies will require large cohorts and comprehensive analytical strategies that consider the substantial multiple-testing burden.
Collapse
|
135
|
DISC1 regulates lactate metabolism in astrocytes: implications for psychiatric disorders. Transl Psychiatry 2018; 8:76. [PMID: 29643356 PMCID: PMC5895599 DOI: 10.1038/s41398-018-0123-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 01/11/2018] [Accepted: 02/18/2018] [Indexed: 12/31/2022] Open
Abstract
Our knowledge of how genetic risk variants contribute to psychiatric disease is mainly limited to neurons. However, the mechanisms whereby the same genetic risk factors could affect the physiology of glial cells remain poorly understood. We studied the role of a psychiatric genetic risk factor, Disrupted-In-Schizophrenia-1 (DISC1), in metabolic functions of astrocytes. We evaluated the effects of knockdown of mouse endogenous DISC1 (DISC1-KD) and expression of a dominant-negative, C-terminus truncated human DISC1 (DN-DISC1) on the markers of energy metabolism, including glucose uptake and lactate production, in primary astrocytes and in mice with selective expression of DN-DISC1 in astrocytes. We also assessed the effects of lactate treatment on altered affective behaviors and impaired spatial memory in DN-DISC1 mice. Both DISC1-KD and DN-DISC1 comparably decreased mRNA and protein levels of glucose transporter 4 and glucose uptake by primary astrocytes. Decreased glucose uptake was associated with reduced oxidative phosphorylation and glycolysis as well as diminished lactate production in vitro and in vivo. No significant effects of DISC1 manipulations in astrocytes were observed on expression of the subunits of the electron transport chain complexes or mitofilin, a neuronal DISC1 partner. Lactate treatment rescued the abnormal behaviors in DN-DISC1 male and female mice. Our results suggest that DISC1 may be involved in the regulation of lactate production in astrocytes to support neuronal activity and associated behaviors. Abnormal expression of DISC1 in astrocytes and resulting abnormalities in energy supply may be responsible for aspects of mood and cognitive disorders observed in patients with major psychiatric illnesses.
Collapse
|
136
|
Comparative genomic evidence for the involvement of schizophrenia risk genes in antipsychotic effects. Mol Psychiatry 2018; 23:708-712. [PMID: 28555076 PMCID: PMC5709242 DOI: 10.1038/mp.2017.111] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 03/08/2017] [Accepted: 04/12/2017] [Indexed: 01/14/2023]
Abstract
Genome-wide association studies (GWAS) for schizophrenia have identified over 100 loci encoding >500 genes. It is unclear whether any of these genes, other than dopamine receptor D2, are immediately relevant to antipsychotic effects or represent novel antipsychotic targets. We applied an in vivo molecular approach to this question by performing RNA sequencing of brain tissue from mice chronically treated with the antipsychotic haloperidol or vehicle. We observed significant enrichments of haloperidol-regulated genes in schizophrenia GWAS loci and in schizophrenia-associated biological pathways. Our findings provide empirical support for overlap between genetic variation underlying the pathophysiology of schizophrenia and the molecular effects of a prototypical antipsychotic.
Collapse
|
137
|
Costas J. The highly pleiotropic gene SLC39A8 as an opportunity to gain insight into the molecular pathogenesis of schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2018; 177:274-283. [PMID: 28557351 DOI: 10.1002/ajmg.b.32545] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 03/28/2017] [Indexed: 12/19/2022]
Abstract
There is a long way from the initial discovery of a genome-wide significant signal to mechanistic understanding of the association. Identification of the gene and causal polymorphism usually requires an extensive additional effort. The schizophrenia genome-wide significant locus at 4q24 may be a rare exception to this pattern. As discussed in this review, the association at this locus is most probably driven by a functional missense variant at the metal cations transporter SLC39A8. The variant, rs13107325, is almost exclusive of European populations and is one of the most pleiotropic variants of the genome, being associated at genome-wide significant level with several additional traits, such as body mass index, Crohn's disease, blood pressure related-traits, and serum levels of manganese, N-terminal pro-B-type natriuretic peptide and HDL-cholesterol. SLC39A8 seems to be subject to recent natural selection in Europeans. It is almost ubiquitously expressed and its physiological role is beginning to be elucidated, mainly in relation to immunity. This manuscript presents arguments in favor of the rs13107325 variant as the functional variant responsible for the association of this locus with schizophrenia, reviews the genetic associations with this gene, the evidences of natural selection on the gene, and the known aspects about its structure and physiological functions. Finally, some hypotheses about putative mechanisms for its association with schizophrenia are presented based on this knowledge, including impaired immunity/inflammation, interference with glutamatergic neurotransmission, homeostasis of essential metals in brain, such as iron, zinc or manganese, or neurotoxicity by heavy metals, such as cadmium or lead.
Collapse
Affiliation(s)
- Javier Costas
- Grupo de Xenética Psiquiátrica, Hospital Clínico Universitario de Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago (IDIS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| |
Collapse
|
138
|
Taylor S. Association between COMT Val158Met and psychiatric disorders: A comprehensive meta-analysis. Am J Med Genet B Neuropsychiatr Genet 2018; 177:199-210. [PMID: 28608575 DOI: 10.1002/ajmg.b.32556] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/08/2017] [Accepted: 05/05/2017] [Indexed: 01/26/2023]
Abstract
Catechol-O-methyltransferase (COMT) Val158Met is widely regarded as potentially important for understanding the genetic etiology of many different psychiatric disorders. The present study appears to be the first comprehensive meta-analysis of COMT genetic association studies to cover all psychiatric disorders for which there were available data, published in any language, and with an emphasis on investigating disorder subtypes (defined clinically or by demographic or other variables). Studies were included if they reported one or more datasets (i.e., some studies examined more than one clinical group) in which there were sufficient information to compute effect sizes. A total of 363 datasets were included, consisting of 56,998 cases and 74,668 healthy controls from case control studies, and 2,547 trios from family based studies. Fifteen disorders were included. Attention-deficit hyperactivity disorder and panic disorder were associated with the Val allele for Caucasian samples. Substance-use disorder, defined by DSM-IV criteria, was associated with the Val allele for Asian samples. Bipolar disorder was associated with the Met allele in Asian samples. Obsessive-compulsive disorder tended to be associated with the Met allele only for males. There was suggestive evidence that the Met allele is associated with an earlier age of onset of schizophrenia. Results suggest pleiotropy and underscore the importance of examining subgroups-defined by variables such as age of onset, sex, ethnicity, and diagnostic system-rather than examining disorders as monolithic constructs.
Collapse
Affiliation(s)
- Steven Taylor
- University of British Columbia, British Columbia, Canada
| |
Collapse
|
139
|
Barone L, Barone V, Dellagiulia A, Lionetti F. Testing an Attachment-Based Parenting Intervention-VIPP-FC/A in Adoptive Families with Post-institutionalized Children: Do Maternal Sensitivity and Genetic Markers Count? Front Psychol 2018; 9:156. [PMID: 29515483 PMCID: PMC5826058 DOI: 10.3389/fpsyg.2018.00156] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/30/2018] [Indexed: 01/01/2023] Open
Abstract
This study investigated the effectiveness of a newly integrated version of an intervention targeting adoptive mothers' positive parenting for promoting children's emotional availability, by testing the moderating role of both two maternal genetic polymorphisms (i.e., 5HTTLPR and DRD4-VNTR) and emotional availability-EA on intervention outcomes. Mothers with their children (N = 80; Mage = 42.73 years, SD = 3.79; Mage = 33.18 months, SD = 16.83 months) participated in a RCT testing the Video-Feedback Intervention to Promote Positive Parenting and Sensitive Discipline-VIPP-FC/A effectiveness. Mixed effects regression models showed a significant improvement in mother-child EA for the VIPP-intervention vs. the dummy intervention condition, with a moderating role of maternal EA on children's outcomes. No significant moderating effect was found for the two genetic polymorphisms inquired. Children's and mother's outcomes obtained are discussed.
Collapse
Affiliation(s)
- Lavinia Barone
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Virginia Barone
- Molecular and Developmental Medicine Department, University of Siena, Siena, Italy
| | | | - Francesca Lionetti
- Department of Biological and Experimental Psychology, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
140
|
Zai CC, Maes MS, Tiwari AK, Zai GC, Remington G, Kennedy JL. Genetics of tardive dyskinesia: Promising leads and ways forward. J Neurol Sci 2018; 389:28-34. [PMID: 29502799 DOI: 10.1016/j.jns.2018.02.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/02/2018] [Indexed: 12/23/2022]
Abstract
Tardive dyskinesia (TD) is a potentially irreversible and often debilitating movement disorder secondary to chronic use of dopamine receptor blocking medications. Genetic factors have been implicated in the etiology of TD. We therefore have reviewed the most promising genes associated with TD, including DRD2, DRD3, VMAT2, HSPG2, HTR2A, HTR2C, and SOD2. In addition, we present evidence supporting a role for these genes from preclinical models of TD. The current understanding of the etiogenesis of TD is discussed in the light of the recent approvals of valbenazine and deutetrabenazine, VMAT2 inhibitors, for treating TD.
Collapse
Affiliation(s)
- Clement C Zai
- Neurogenetics Section, Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Canada; Department of Psychiatry, University of Toronto, Canada; Institute of Medical Science, University of Toronto, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Canada.
| | - Miriam S Maes
- Neurogenetics Section, Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Canada
| | - Arun K Tiwari
- Neurogenetics Section, Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Canada; Department of Psychiatry, University of Toronto, Canada
| | - Gwyneth C Zai
- Neurogenetics Section, Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Canada; Department of Psychiatry, University of Toronto, Canada
| | - Gary Remington
- Neurogenetics Section, Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Canada; Department of Psychiatry, University of Toronto, Canada; Institute of Medical Science, University of Toronto, Canada
| | - James L Kennedy
- Neurogenetics Section, Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Canada; Department of Psychiatry, University of Toronto, Canada; Institute of Medical Science, University of Toronto, Canada.
| |
Collapse
|
141
|
Hartung H, Cichon N, De Feo V, Riemann S, Schildt S, Lindemann C, Mulert C, Gogos JA, Hanganu-Opatz IL. From Shortage to Surge: A Developmental Switch in Hippocampal-Prefrontal Coupling in a Gene-Environment Model of Neuropsychiatric Disorders. Cereb Cortex 2018; 26:4265-4281. [PMID: 27613435 PMCID: PMC5066837 DOI: 10.1093/cercor/bhw274] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/10/2016] [Indexed: 12/21/2022] Open
Abstract
Cognitive deficits represent a major burden of neuropsychiatric disorders and result in part from abnormal communication within hippocampal–prefrontal circuits. While it has been hypothesized that this network dysfunction arises during development, long before the first clinical symptoms, experimental evidence is still missing. Here, we show that pre-juvenile mice mimicking genetic and environmental risk factors of disease (dual-hit GE mice) have poorer recognition memory that correlates with augmented coupling by synchrony and stronger directed interactions between prefrontal cortex and hippocampus. The network dysfunction emerges already during neonatal development, yet it initially consists in a diminished hippocampal theta drive and consequently, a weaker and disorganized entrainment of local prefrontal circuits in discontinuous oscillatory activity in dual-hit GE mice when compared with controls. Thus, impaired maturation of functional communication within hippocampal–prefrontal networks switching from hypo- to hyper-coupling may represent a mechanism underlying the pathophysiology of cognitive deficits in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Henrike Hartung
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Laboratory of Neurobiology, Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland
| | - Nicole Cichon
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Vito De Feo
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Laboratory of Neural Computation, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| | - Stephanie Riemann
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Current address: German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Sandra Schildt
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christoph Lindemann
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christoph Mulert
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Joseph A Gogos
- Department of Neuroscience, Columbia University, New York, NY 10032, USA.,Department of Physiology, Columbia University, New York, NY 10032, USA
| | - Ileana L Hanganu-Opatz
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
142
|
Arslan A. Mapping the Schizophrenia Genes by Neuroimaging: The Opportunities and the Challenges. Int J Mol Sci 2018; 19:ijms19010219. [PMID: 29324666 PMCID: PMC5796168 DOI: 10.3390/ijms19010219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/05/2018] [Accepted: 01/07/2018] [Indexed: 12/18/2022] Open
Abstract
Schizophrenia (SZ) is a heritable brain disease originating from a complex interaction of genetic and environmental factors. The genes underpinning the neurobiology of SZ are largely unknown but recent data suggest strong evidence for genetic variations, such as single nucleotide polymorphisms, making the brain vulnerable to the risk of SZ. Structural and functional brain mapping of these genetic variations are essential for the development of agents and tools for better diagnosis, treatment and prevention of SZ. Addressing this, neuroimaging methods in combination with genetic analysis have been increasingly used for almost 20 years. So-called imaging genetics, the opportunities of this approach along with its limitations for SZ research will be outlined in this invited paper. While the problems such as reproducibility, genetic effect size, specificity and sensitivity exist, opportunities such as multivariate analysis, development of multisite consortia for large-scale data collection, emergence of non-candidate gene (hypothesis-free) approach of neuroimaging genetics are likely to contribute to a rapid progress for gene discovery besides to gene validation studies that are related to SZ.
Collapse
Affiliation(s)
- Ayla Arslan
- Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnica cesta, 15 Ilidza, Sarajevo 71210, Bosnia and Herzegovina.
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul 34662, Turkey.
| |
Collapse
|
143
|
Abstract
Imaging genetics is a research methodology studying the effect of genetic variation on brain structure, function, behavior, and risk for psychopathology. Since the early 2000s, imaging genetics has been increasingly used in the research of schizophrenia (SZ). SZ is a severe mental disorder with no precise knowledge of its underlying neurobiology, however, new genetic and neurobiological data generate a climate for new avenues. The accumulating data of genome wide association studies (GWAS) continuously decode SZ risk genes. Global neuroimaging consortia produce collections of brain phenotypes from tens of thousands of people. In this context, imaging genetics will be strategically important both for the validation and discovery of SZ related findings. Thus, the study of GWAS supported risk variants as candidate genes to validate by neuroimaging is one trend. The study of epigenetic differences in relation to variations of brain phenotypes and the study of large scale multivariate analysis of genome wide and brain wide associations are other trends. While these studies hold a big potential for understanding the neurobiology of SZ, the problem of reproducibility appears as a major challenge, which requires standardizations in study designs and compensations of methodological limitations such as sensitivity and specificity. On the other hand, advancements of neuroimaging, optical and electron microscopy along with the use of genetically encoded fluorescent probes and robust statistical approaches will not only catalyze integrative methodologies but also will help better design the imaging genetics studies. In this invited paper, I will discuss the current perspective of imaging genetics and emerging opportunities of SZ research.
Collapse
Affiliation(s)
- Ayla Arslan
- Faculty of Engineering and Natural Sciences, Department of Genetics and Bioengineering, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina; Faculty of Engineering and Natural Sciences, Department of Molecular Biology and Genetics, Uskudar University, Istanbul, Turkey.
| |
Collapse
|
144
|
Latendresse SJ, Musci R, Maher BS. Critical Issues in the Inclusion of Genetic and Epigenetic Information in Prevention and Intervention Trials. PREVENTION SCIENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR PREVENTION RESEARCH 2018; 19:58-67. [PMID: 28409280 PMCID: PMC5640466 DOI: 10.1007/s11121-017-0785-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human genetic research in the past decade has generated a wealth of data from the genome-wide association scan era, much of which is catalogued and freely available. These data will typically test the relationship between a single nucleotide variant or polymorphism (SNP) and some outcome, disease, or trait. Ongoing investigations will yield a similar wealth of data regarding epigenetic phenomena. These data will typically test the relationship between DNA methylation at a single genomic location/region and some outcome. Most of these findings will be the result of cross-sectional investigations typically using ascertained cases and controls. Consequently, most methodological consideration focuses on methods appropriate for simple case-control comparisons. It is expected that a growing number of investigators with longitudinal experimental prevention or intervention cohorts will also measure genetic and epigenetic indicators as part of their investigations, harvesting the wealth of information generated by the genome-wide association study (GWAS) era to allow for targeted hypothesis testing in the next generation of prevention and intervention trials. Herein, we discuss appropriate quality control and statistical modelling of genetic, polygenic, and epigenetic measures in longitudinal models. We specifically discuss quality control, population stratification, genotype imputation, pathway approaches, and proper modelling of an interaction between a specific genetic variant and an environment variable (GxE interaction).
Collapse
Affiliation(s)
- Shawn J Latendresse
- Department of Psychology and Neuroscience, Baylor University, One Bear Place #97334, Waco, TX, 76798, USA.
| | - Rashelle Musci
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway Ave, Baltimore, MD, 21205, USA
| | - Brion S Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway Ave, Baltimore, MD, 21205, USA.
| |
Collapse
|
145
|
Suvisaari J, Mantere O, Keinänen J, Mäntylä T, Rikandi E, Lindgren M, Kieseppä T, Raij TT. Is It Possible to Predict the Future in First-Episode Psychosis? Front Psychiatry 2018; 9:580. [PMID: 30483163 PMCID: PMC6243124 DOI: 10.3389/fpsyt.2018.00580] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/23/2018] [Indexed: 12/26/2022] Open
Abstract
The outcome of first-episode psychosis (FEP) is highly variable, ranging from early sustained recovery to antipsychotic treatment resistance from the onset of illness. For clinicians, a possibility to predict patient outcomes would be highly valuable for the selection of antipsychotic treatment and in tailoring psychosocial treatments and psychoeducation. This selective review summarizes current knowledge of prognostic markers in FEP. We sought potential outcome predictors from clinical and sociodemographic factors, cognition, brain imaging, genetics, and blood-based biomarkers, and we considered different outcomes, like remission, recovery, physical comorbidities, and suicide risk. Based on the review, it is currently possible to predict the future for FEP patients to some extent. Some clinical features-like the longer duration of untreated psychosis (DUP), poor premorbid adjustment, the insidious mode of onset, the greater severity of negative symptoms, comorbid substance use disorders (SUDs), a history of suicide attempts and suicidal ideation and having non-affective psychosis-are associated with a worse outcome. Of the social and demographic factors, male gender, social disadvantage, neighborhood deprivation, dysfunctional family environment, and ethnicity may be relevant. Treatment non-adherence is a substantial risk factor for relapse, but a small minority of patients with acute onset of FEP and early remission may benefit from antipsychotic discontinuation. Cognitive functioning is associated with functional outcomes. Brain imaging currently has limited utility as an outcome predictor, but this may change with methodological advancements. Polygenic risk scores (PRSs) might be useful as one component of a predictive tool, and pharmacogenetic testing is already available and valuable for patients who have problems in treatment response or with side effects. Most blood-based biomarkers need further validation. None of the currently available predictive markers has adequate sensitivity or specificity used alone. However, personalized treatment of FEP will need predictive tools. We discuss some methodologies, such as machine learning (ML), and tools that could lead to the improved prediction and clinical utility of different prognostic markers in FEP. Combination of different markers in ML models with a user friendly interface, or novel findings from e.g., molecular genetics or neuroimaging, may result in computer-assisted clinical applications in the near future.
Collapse
Affiliation(s)
- Jaana Suvisaari
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Outi Mantere
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Psychiatry, McGill University, Montreal, QC, Canada.,Bipolar Disorders Clinic, Douglas Mental Health University Institute, Montreal, QC, Canada.,Department of Psychiatry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jaakko Keinänen
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Psychiatry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Teemu Mäntylä
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Neuroscience and Biomedical Engineering, and Advanced Magnetic Imaging Center, Aalto NeuroImaging, Aalto University School of Science, Espoo, Finland.,Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Eva Rikandi
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Neuroscience and Biomedical Engineering, and Advanced Magnetic Imaging Center, Aalto NeuroImaging, Aalto University School of Science, Espoo, Finland.,Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Maija Lindgren
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Tuula Kieseppä
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Psychiatry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tuukka T Raij
- Mental Health Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of Neuroscience and Biomedical Engineering, and Advanced Magnetic Imaging Center, Aalto NeuroImaging, Aalto University School of Science, Espoo, Finland
| |
Collapse
|
146
|
Investigation of previously implicated genetic variants in chronic tic disorders: a transmission disequilibrium test approach. Eur Arch Psychiatry Clin Neurosci 2018; 268:301-316. [PMID: 28555406 PMCID: PMC5708161 DOI: 10.1007/s00406-017-0808-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 05/17/2017] [Indexed: 12/25/2022]
Abstract
Genetic studies in Tourette syndrome (TS) are characterized by scattered and poorly replicated findings. We aimed to replicate findings from candidate gene and genome-wide association studies (GWAS). Our cohort included 465 probands with chronic tic disorder (93% TS) and both parents from 412 families (some probands were siblings). We assessed 75 single nucleotide polymorphisms (SNPs) in 465 parent-child trios; 117 additional SNPs in 211 trios; and 4 additional SNPs in 254 trios. We performed SNP and gene-based transmission disequilibrium tests and compared nominally significant SNP results with those from a large independent case-control cohort. After quality control 71 SNPs were available in 371 trios; 112 SNPs in 179 trios; and 3 SNPs in 192 trios. 17 were candidate SNPs implicated in TS and 2 were implicated in obsessive-compulsive disorder (OCD) or autism spectrum disorder (ASD); 142 were tagging SNPs from eight monoamine neurotransmitter-related genes (including dopamine and serotonin); 10 were top SNPs from TS GWAS; and 13 top SNPs from attention-deficit/hyperactivity disorder, OCD, or ASD GWAS. None of the SNPs or genes reached significance after adjustment for multiple testing. We observed nominal significance for the candidate SNPs rs3744161 (TBCD) and rs4565946 (TPH2) and for five tagging SNPs; none of these showed significance in the independent cohort. Also, SLC1A1 in our gene-based analysis and two TS GWAS SNPs showed nominal significance, rs11603305 (intergenic) and rs621942 (PICALM). We found no convincing support for previously implicated genetic polymorphisms. Targeted re-sequencing should fully appreciate the relevance of candidate genes.
Collapse
|
147
|
Gassó P, Mas S, Rodríguez N, Boloc D, García-Cerro S, Bernardo M, Lafuente A, Parellada E. Microarray gene-expression study in fibroblast and lymphoblastoid cell lines from antipsychotic-naïve first-episode schizophrenia patients. J Psychiatr Res 2017; 95:91-101. [PMID: 28822801 DOI: 10.1016/j.jpsychires.2017.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/25/2017] [Accepted: 08/04/2017] [Indexed: 12/16/2022]
Abstract
Schizophrenia (SZ) is a chronic psychiatric disorder whose onset of symptoms occurs in late adolescence and early adulthood. The etiology is complex and involves important gene-environment interactions. Microarray gene-expression studies on SZ have identified alterations in several biological processes. The heterogeneity in the results can be attributed to the use of different sample types and other important confounding factors including age, illness chronicity and antipsychotic exposure. The aim of the present microarray study was to analyze, for the first time to our knowledge, differences in gene expression profiles in 18 fibroblast (FCLs) and 14 lymphoblastoid cell lines (LCLs) from antipsychotic-naïve first-episode schizophrenia (FES) patients and healthy controls. We used an analytical approach based on protein-protein interaction network construction and functional annotation analysis to identify the biological processes that are altered in SZ. Significant differences in the expression of 32 genes were found when LCLs were assessed. The network and gene set enrichment approach revealed the involvement of similar biological processes in FCLs and LCLs, including apoptosis and related biological terms such as cell cycle, autophagy, cytoskeleton organization and response to stress and stimulus. Metabolism and other processes, including signal transduction, kinase activity and phosphorylation, were also identified. These results were replicated in two independent cohorts using the same analytical approach. This provides more evidence for altered apoptotic processes in antipsychotic-naïve FES patients and other important biological functions such as cytoskeleton organization and metabolism. The convergent results obtained in both peripheral cell models support their usefulness for transcriptome studies on SZ.
Collapse
Affiliation(s)
- Patricia Gassó
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Sergi Mas
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | | | - Daniel Boloc
- Dept. of Basic Clinical Practice, University of Barcelona, Spain
| | | | - Miquel Bernardo
- Barcelona Clínic Schizophrenia Unit (BCSU), Neuroscience Institute, Hospital Clínic de Barcelona, Spain; Dept. of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Amalia Lafuente
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Eduard Parellada
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Barcelona Clínic Schizophrenia Unit (BCSU), Neuroscience Institute, Hospital Clínic de Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| |
Collapse
|
148
|
Johnson EC, Border R, Melroy-Greif WE, de Leeuw C, Ehringer MA, Keller MC. No Evidence That Schizophrenia Candidate Genes Are More Associated With Schizophrenia Than Noncandidate Genes. Biol Psychiatry 2017; 82:702-708. [PMID: 28823710 PMCID: PMC5643230 DOI: 10.1016/j.biopsych.2017.06.033] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 06/12/2017] [Accepted: 06/28/2017] [Indexed: 01/12/2023]
Abstract
BACKGROUND A recent analysis of 25 historical candidate gene polymorphisms for schizophrenia in the largest genome-wide association study conducted to date suggested that these commonly studied variants were no more associated with the disorder than would be expected by chance. However, the same study identified other variants within those candidate genes that demonstrated genome-wide significant associations with schizophrenia. As such, it is possible that variants within historic schizophrenia candidate genes are associated with schizophrenia at levels above those expected by chance, even if the most-studied specific polymorphisms are not. METHODS The present study used association statistics from the largest schizophrenia genome-wide association study conducted to date as input to a gene set analysis to investigate whether variants within schizophrenia candidate genes are enriched for association with schizophrenia. RESULTS As a group, variants in the most-studied candidate genes were no more associated with schizophrenia than were variants in control sets of noncandidate genes. While a small subset of candidate genes did appear to be significantly associated with schizophrenia, these genes were not particularly noteworthy given the large number of more strongly associated noncandidate genes. CONCLUSIONS The history of schizophrenia research should serve as a cautionary tale to candidate gene investigators examining other phenotypes: our findings indicate that the most investigated candidate gene hypotheses of schizophrenia are not well supported by genome-wide association studies, and it is likely that this will be the case for other complex traits as well.
Collapse
Affiliation(s)
- Emma C. Johnson
- Department of Psychology and Neuroscience, University of Colorado at Boulder, USA,Institute for Behavioral Genetics, University of Colorado at Boulder, USA,Correspondence concerning this article should be addressed to Emma C. Johnson, Institute of Behavioral Genetics, 1480 30th Street, Boulder, CO, 80303.
| | - Richard Border
- Department of Psychology and Neuroscience, University of Colorado at Boulder, USA,Institute for Behavioral Genetics, University of Colorado at Boulder, USA
| | | | - Christiaan de Leeuw
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research/VU University Amsterdam, Amsterdam 1081 HV, Netherlands,Institute for Computing and Information Sciences, Radboud University Nijmegen, Nijmegen 6525 EC, Netherlands
| | - Marissa A. Ehringer
- Institute for Behavioral Genetics, University of Colorado at Boulder, USA,Deparment of Integrative Physiology, University of Colorado at Boulder, USA
| | - Matthew C. Keller
- Department of Psychology and Neuroscience, University of Colorado at Boulder, USA,Institute for Behavioral Genetics, University of Colorado at Boulder, USA
| |
Collapse
|
149
|
Sullivan PF. How Good Were Candidate Gene Guesses in Schizophrenia Genetics? Biol Psychiatry 2017; 82:696-697. [PMID: 29031917 PMCID: PMC6950644 DOI: 10.1016/j.biopsych.2017.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Patrick F Sullivan
- Departments of Genetics and Psychiatry, University of North Carolina at Chapel Hill, North Carolina; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
150
|
Harari JH, Díaz-Caneja CM, Janssen J, Martínez K, Arias B, Arango C. The association between gene variants and longitudinal structural brain changes in psychosis: a systematic review of longitudinal neuroimaging genetics studies. NPJ SCHIZOPHRENIA 2017; 3:40. [PMID: 29093492 PMCID: PMC5665946 DOI: 10.1038/s41537-017-0036-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/18/2017] [Accepted: 08/29/2017] [Indexed: 12/18/2022]
Abstract
Evidence suggests that genetic variation might influence structural brain alterations in psychotic disorders. Longitudinal genetic neuroimaging (G-NI) studies are designed to assess the association between genetic variants, disease progression and brain changes. There is a paucity of reviews of longitudinal G-NI studies in psychotic disorders. A systematic search of PubMed from inception until November 2016 was conducted to identify longitudinal G-NI studies examining the link between Magnetic Resonance Imaging (MRI) and Diffusion Tensor Imaging (DTI)-based brain measurements and specific gene variants (SNPs, microsatellites, haplotypes) in patients with psychosis. Eleven studies examined seven genes: BDNF, COMT, NRG1, DISC1, CNR1, GAD1, and G72. Eight of these studies reported at least one association between a specific gene variant and longitudinal structural brain changes. Genetic variants associated with longitudinal brain volume or cortical thickness loss included a 4-marker haplotype in G72, a microsatellite and a SNP in NRG1, and individual SNPs in DISC1, CNR1, BDNF, COMT and GAD1. Associations between genotype and progressive brain changes were most frequently observed in frontal regions, with five studies reporting significant interactions. Effect sizes for significant associations were generally of small or intermediate magnitude (Cohen’s d < 0.8). Only two genes (BDNF and NRG1) were assessed in more than one study, with great heterogeneity of the results. Replication studies and studies exploring additional genetic variants identified by large-scale genetic analysis are warranted to further ascertain the role of genetic variants in longitudinal brain changes in psychosis.
Collapse
Affiliation(s)
- Julia H Harari
- Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, CIBERSAM, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), School of Medicine, Universidad Complutense, Madrid, Spain.,University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Covadonga M Díaz-Caneja
- Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, CIBERSAM, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Joost Janssen
- Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, CIBERSAM, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), School of Medicine, Universidad Complutense, Madrid, Spain.,Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kenia Martínez
- Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, CIBERSAM, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Bárbara Arias
- Zoology and Biological Anthropology Unit. Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals. IBUB., Faculty of Biology, Universitat de Barcelona, Barcelona, Spain. .,CIBERSAM (Centro de Investigación Biomédica en Red de Salud Mental), Instituto de Salud Carlos III, Madrid, Spain.
| | - Celso Arango
- Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, CIBERSAM, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), School of Medicine, Universidad Complutense, Madrid, Spain.
| |
Collapse
|