Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
medRxiv ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38712091

ABSTRACT

Obsessive-compulsive disorder (OCD) affects ~1% of the population and exhibits a high SNP-heritability, yet previous genome-wide association studies (GWAS) have provided limited information on the genetic etiology and underlying biological mechanisms of the disorder. We conducted a GWAS meta-analysis combining 53,660 OCD cases and 2,044,417 controls from 28 European-ancestry cohorts revealing 30 independent genome-wide significant SNPs and a SNP-based heritability of 6.7%. Separate GWAS for clinical, biobank, comorbid, and self-report sub-groups found no evidence of sample ascertainment impacting our results. Functional and positional QTL gene-based approaches identified 249 significant candidate risk genes for OCD, of which 25 were identified as putatively causal, highlighting WDR6, DALRD3, CTNND1 and genes in the MHC region. Tissue and single-cell enrichment analyses highlighted hippocampal and cortical excitatory neurons, along with D1- and D2-type dopamine receptor-containing medium spiny neurons, as playing a role in OCD risk. OCD displayed significant genetic correlations with 65 out of 112 examined phenotypes. Notably, it showed positive genetic correlations with all included psychiatric phenotypes, in particular anxiety, depression, anorexia nervosa, and Tourette syndrome, and negative correlations with a subset of the included autoimmune disorders, educational attainment, and body mass index.. This study marks a significant step toward unraveling its genetic landscape and advances understanding of OCD genetics, providing a foundation for future interventions to address this debilitating disorder.

3.
medRxiv ; 2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37461564

ABSTRACT

Diagnostic criteria for major depressive disorder allow for heterogeneous symptom profiles but genetic analysis of major depressive symptoms has the potential to identify clinical and aetiological subtypes. There are several challenges to integrating symptom data from genetically-informative cohorts, such as sample size differences between clinical and community cohorts and various patterns of missing data. We conducted genome-wide association studies of major depressive symptoms in three clinical cohorts that were enriched for affected participants (Psychiatric Genomics Consortium, Australian Genetics of Depression Study, Generation Scotland) and three community cohorts (Avon Longitudinal Study of Parents and Children, Estonian Biobank, and UK Biobank). We fit a series of confirmatory factor models with factors that accounted for how symptom data was sampled and then compared alternative models with different symptom factors. The best fitting model had a distinct factor for Appetite/Weight symptoms and an additional measurement factor that accounted for missing data patterns in the community cohorts (use of Depression and Anhedonia as gating symptoms). The results show the importance of assessing the directionality of symptoms (such as hypersomnia versus insomnia) and of accounting for study and measurement design when meta-analysing genetic association data.

4.
Psychiatry Res ; 326: 115343, 2023 08.
Article in English | MEDLINE | ID: mdl-37473490

ABSTRACT

Anxiety disorders are a group of prevalent and heritable neuropsychiatric diseases. We previously conducted a genome-wide association study (GWAS) which identified genomic loci associated with anxiety; however, the biological consequences underlying the genetic associations are largely unknown. Integrating GWAS and functional genomic data may improve our understanding of the genetic effects on intermediate molecular phenotypes such as gene expression. This can provide an opportunity for the discovery of drug targets for anxiety via drug repurposing. We used the GWAS summary statistics to determine putative causal genes for anxiety using MAGMA and colocalization analyses. A transcriptome-wide association study was conducted to identify genes with differential genetically regulated levels of gene expression in human brain tissue. The genes were integrated with a large drug-gene expression database (Connectivity Map), discovering compounds that are predicted to "normalise" anxiety-associated expression changes. The study identified 64 putative causal genes associated with anxiety (35 genes upregulated; 29 genes downregulated). Drug mechanisms adrenergic receptor agonists, sigma receptor agonists, and glutamate receptor agonists gene targets were enriched in anxiety-associated genetic signal and exhibited an opposing effect on the anxiety-associated gene expression signature. The significance of the project demonstrated genetic links for novel drug candidates to potentially advance anxiety therapeutics.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Drug Repositioning , Transcriptome , Anxiety/drug therapy , Anxiety/genetics , Anxiety Disorders/drug therapy , Anxiety Disorders/genetics , Polymorphism, Single Nucleotide
5.
EBioMedicine ; 92: 104615, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37201334

ABSTRACT

BACKGROUND: Primary open-angle glaucoma (POAG) is an optic neuropathy characterized by progressive degeneration of the optic nerve that leads to irreversible visual impairment. Multiple epidemiological studies suggest an association between POAG and major neurodegenerative disorders (Alzheimer's disease, amyotrophic lateral sclerosis, frontotemporal dementia, and Parkinson's disease). However, the nature of the overlap between neurodegenerative disorders, brain morphology and glaucoma remains inconclusive. METHOD: In this study, we performed a comprehensive assessment of the genetic and causal relationship between POAG and neurodegenerative disorders, leveraging genome-wide association data from studies of magnetic resonance imaging of the brain, POAG, and four major neurodegenerative disorders. FINDINGS: This study found a genetic overlap and causal relationship between POAG and its related phenotypes (i.e., intraocular pressure and optic nerve morphology traits) and brain morphology in 19 regions. We also identified 11 loci with a significant local genetic correlation and a high probability of sharing the same causal variant between neurodegenerative disorders and POAG or its related phenotypes. Of interest, a region on chromosome 17 corresponding to MAPT, a well-known risk locus for Alzheimer's and Parkinson's disease, was shared between POAG, optic nerve degeneration traits, and Alzheimer's and Parkinson's diseases. Despite these local genetic overlaps, we did not identify strong evidence of a causal association between these neurodegenerative disorders and glaucoma. INTERPRETATION: Our findings indicate a distinctive and likely independent neurodegenerative process for POAG involving several brain regions although several POAG or optic nerve degeneration risk loci are shared with neurodegenerative disorders, consistent with a pleiotropic effect rather than a causal relationship between these traits. FUNDING: PG was supported by an NHMRC Investigator Grant (#1173390), SM by an NHMRC Senior Research Fellowship and an NHMRC Program Grant (APP1150144), DM by an NHMRC Fellowship, LP is funded by the NEIEY015473 and EY032559 grants, SS is supported by an NIH-Oxford Cambridge Fellowship and NIH T32 grant (GM136577), APK is supported by a UK Research and Innovation Future Leaders Fellowship, an Alcon Research Institute Young Investigator Award and a Lister Institute for Preventive Medicine Award.


Subject(s)
Alzheimer Disease , Glaucoma, Open-Angle , Glaucoma , Neurodegenerative Diseases , Parkinson Disease , Humans , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/pathology , Genome-Wide Association Study , Parkinson Disease/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Glaucoma/genetics , Brain/diagnostic imaging , Brain/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology
6.
Psychiatry Res ; 321: 115101, 2023 03.
Article in English | MEDLINE | ID: mdl-36774750

ABSTRACT

BACKGROUND: Traumatic experiences are associated with increased risk for major depressive disorder (MDD). This study sought to determine the extent that trauma exposure, depression polygenic risk scores (PRS), and their interaction are associated with MDD and individual depression symptoms. METHODS: Data from 102,182 individuals from the large-scale UK Biobank population cohort was analysed. A series of regression analyses were conducted to estimate the association between trauma, depression PRS and 1) current depression, 2) lifetime MDD case-control status, 3) nine individual current depressive symptoms, and 4) thirteen individual symptoms experienced during a major depressive episode. Additive and multiplicative PRS-by-trauma interactions were also assessed. RESULTS: Trauma and depression PRS were significantly associated with both current depression and lifetime MDD. A positive, additive interaction effect was observed on depression, but multiplicative interactions were not significant. Trauma exposure and depression PRS were associated with specific patterns of depression symptoms; Trauma was associated with low self-esteem, suicidal ideation, and atypical (but not typical) neurovegetative symptoms. Additive interaction effects were observed on six out of nine current depressive symptoms. CONCLUSIONS: Trauma exposure and genetic predisposition to depression may lead to particular symptomatology, which may contribute to the extreme clinical heterogeneity observed in individuals with major depression.


Subject(s)
Depressive Disorder, Major , Humans , Depressive Disorder, Major/diagnosis , Depression , Genetic Predisposition to Disease , Risk Factors , Regression Analysis , Genome-Wide Association Study
7.
Nat Genet ; 54(10): 1457-1465, 2022 10.
Article in English | MEDLINE | ID: mdl-36138228

ABSTRACT

Genome-wide association studies have identified hundreds of robust genetic associations underlying psychiatric disorders and provided important biological insights into disease onset and progression. There is optimism that genetic findings will pave the way to precision psychiatry by facilitating the development of more effective treatments and the identification of groups of patients that these treatments should be targeted toward. However, there are several challenges that must be addressed before genetic findings can be translated into the clinic. In this Perspective, we highlight ten challenges for the field of psychiatric genetics, focused on the robust and generalizable detection of genetic risk factors, improved definition and assessment of psychopathology and achieving better clinical indicators. We discuss recent advancements in the field that will improve the explanatory and predictive power of genetic data and ultimately contribute to improving the management and treatment of patients with a psychiatric disorder.


Subject(s)
Mental Disorders , Psychiatry , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Mental Disorders/genetics
8.
Neurobiol Aging ; 119: 127-135, 2022 11.
Article in English | MEDLINE | ID: mdl-35989212

ABSTRACT

Alzheimer's disease (AD) is predicted to affect 132 million people by 2050. Targeting modifiable lifestyle risk factors that are associated with an increased risk of AD could prevent a large proportion of dementia cases, allowing people to reach the end of their life dementia free. However, evidence obtained from the observational studies does not take into account how risk factors are correlated with one another, and whether they causally contribute to increased AD risk. In this study, we determine whether the relationship between previously speculated AD risk factors and AD susceptibility is consistent with causality using large-scale genetic data. We focus on educational attainment (EA), intelligence and household income which have been previously shown to be causally associated with AD. Using GWAS-by-subtraction and Multivariable Mendelian Randomization we show that of these, only the cognitive component of EA (intelligence) is independently causally associated with AD. This work has ramifications for the modifiability of lifestyle risk factors for AD.


Subject(s)
Alzheimer Disease , Mendelian Randomization Analysis , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Educational Status , Genome-Wide Association Study , Humans , Intelligence/genetics , Polymorphism, Single Nucleotide , Risk Factors
9.
Schizophr Bull ; 48(6): 1318-1326, 2022 11 18.
Article in English | MEDLINE | ID: mdl-35925031

ABSTRACT

BACKGROUND AND HYPOTHESIS: The nature of the robust association between cannabis use and schizophrenia remains undetermined. Plausible hypotheses explaining this relationship include the premise that cannabis use causes schizophrenia, increased liability for schizophrenia increases the risk of cannabis use initiation (eg, self-medication), or the bidirectional causal hypothesis where both factors play a role in the development of the other. Alternatively, factors that confound the relationship between schizophrenia and cannabis use may explain their association. Externalizing behaviors are related to both schizophrenia and cannabis use and may influence their relationship. STUDY DESIGN: This study aimed to evaluate whether externalizing behaviors influence the genetic relationship between cannabis use and schizophrenia. We conducted a multivariate genome-wide association analysis of 6 externalizing behaviors in order to construct a genetic latent factor of the externalizing spectrum. Genomic structural equation modeling was used to evaluate the influence of externalizing behaviors on the genetic relationship between cannabis use and schizophrenia. RESULTS: We found that externalizing behaviors partially explained the association between cannabis use and schizophrenia by up to 42%. CONCLUSIONS: This partial explanation of the association by externalizing behaviors suggests that there may be other unidentified confounding factors, alongside a possible direct association between schizophrenia and cannabis use. Future studies should aim to identify further confounding factors to accurately explain the relationship between cannabis use and schizophrenia.


Subject(s)
Cannabis , Marijuana Abuse , Schizophrenia , Humans , Schizophrenia/epidemiology , Schizophrenia/genetics , Genome-Wide Association Study , Risk Factors , Marijuana Abuse/epidemiology , Marijuana Abuse/genetics
10.
Biol Psychiatry ; 92(7): 583-591, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35525699

ABSTRACT

BACKGROUND: Global genetic correlation analysis has provided valuable insight into the shared genetic basis between psychiatric and substance use disorders. However, little is known about which regions disproportionately contribute to the global correlation. METHODS: We used Local Analysis of [co]Variant Annotation to calculate bivariate local genetic correlations across 2495 approximately equal-sized, semi-independent genomic regions for 20 psychiatric and substance use phenotypes. We performed a transcriptome-wide association study using expression weights from the prefrontal cortex to identify risk genes for each phenotype, followed by probabilistic fine-mapping to prioritize credible causal genes within each bivariate locus. RESULTS: We detected 80 significant (p < 2.08 × 10-6) bivariate local genetic correlations across 61 loci. The expression effect directions for risk genes within each bivariate locus were largely consistent with the local correlation coefficients, suggesting that genetically regulated gene expression may be used in the functional interpretation of local genetic correlations. Probabilistic fine-mapping identified several genes that may drive pleiotropic mechanisms for genetically correlated phenotypes. For example, we confirmed a local genetic correlation between schizophrenia and smoking behavior at 15q25 and prioritized PSMA4 as the most credible gene candidate underlying both phenotypes. CONCLUSIONS: Our study reveals previously unreported local bivariate genetic correlations between psychiatric and substance use phenotypes, which we fine-mapped to identify shared credible causal genes underlying genetically correlated phenotypes.


Subject(s)
Schizophrenia , Substance-Related Disorders , Genetic Predisposition to Disease , Genome-Wide Association Study , Genomics , Humans , Phenotype , Polymorphism, Single Nucleotide/genetics , Schizophrenia/genetics , Substance-Related Disorders/genetics
11.
Hum Mol Genet ; 31(17): 2887-2898, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35394011

ABSTRACT

Depression is one of the most common mental health disorders and one of the top causes of disability throughout the world. The present study sought to identify putative causal associations between depression and hundreds of complex human traits through a genome-wide screening of genetic data and a hypothesis-free approach. We leveraged genome-wide association studies summary statistics for depression and 1504 complex traits and investigated potential causal relationships using the latent causal variable method. We identified 559 traits genetically correlated with depression risk at FDR < 5%. Of these, 46 were putative causal genetic determinants of depression, including lifestyle factors, diseases of the nervous system, respiratory disorders, diseases of the musculoskeletal system, traits related to the health of the gastrointestinal system, obesity, vitamin D levels and the use of prescription medications, among others. No phenotypes were identified as potential outcomes of depression. Our results suggest that genetic liability to multiple complex traits may contribute to a higher risk for depression. In particular, we show a putative causal genetic effect of pain, obesity and inflammation on depression. These findings provide novel insights into the potential causal determinants of depression and should be interpreted as testable hypotheses for future studies to confirm, which may facilitate the design of new prevention strategies to reduce depression's burden.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Depression/genetics , Genome-Wide Association Study/methods , Humans , Obesity/genetics , Phenomics , Polymorphism, Single Nucleotide/genetics
12.
Eur J Hum Genet ; 30(5): 560-566, 2022 05.
Article in English | MEDLINE | ID: mdl-35217801

ABSTRACT

Genome-wide association studies (GWASs) have identified thousands of risk loci for psychiatric and substance use phenotypes, however the biological consequences of these loci remain largely unknown. We performed a transcriptome-wide association study of 10 psychiatric disorders and 6 substance use phenotypes (GWAS sample size range, N = 9725-807,553) using expression quantitative trait loci data from 532 prefrontal cortex samples. We estimated the correlation of genetically regulated expression between phenotype pairs, and compared the results with the genetic correlations. We identified 393 genes with at least one significant phenotype association, comprising 458 significant associations across 16 phenotypes. Overall, the transcriptomic correlations for phenotype pairs were significantly higher than the respective genetic correlations. For example, attention deficit hyperactivity disorder and autism spectrum disorder, both childhood developmental disorders, had significantly higher transcriptomic correlation (r = 0.84) than genetic correlation (r = 0.35). Finally, we tested the enrichment of phenotype-associated genes in gene co-expression networks built from human prefrontal cortex samples. Phenotype-associated genes were enriched in multiple gene co-expression modules and the implicated modules contained genes involved in mRNA splicing and glutamatergic receptors, among others. Together, our results highlight the utility of gene expression data in the understanding of functional gene mechanisms underlying psychiatric disorders and substance use phenotypes.


Subject(s)
Autism Spectrum Disorder , Substance-Related Disorders , Autism Spectrum Disorder/genetics , Child , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Phenotype , Polymorphism, Single Nucleotide , Substance-Related Disorders/genetics , Transcriptome
13.
JAMA Psychiatry ; 78(10): 1152-1160, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34379077

ABSTRACT

Importance: Genetic studies with broad definitions of depression may not capture genetic risk specific to major depressive disorder (MDD), raising questions about how depression should be operationalized in future genetic studies. Objective: To use a large, well-phenotyped single study of MDD to investigate how different definitions of depression used in genetic studies are associated with estimation of MDD and phenotypes of MDD, using polygenic risk scores (PRSs). Design, Setting, and Participants: In this case-control polygenic risk score analysis, patients meeting diagnostic criteria for a diagnosis of MDD were drawn from the Australian Genetics of Depression Study, a cross-sectional, population-based study of depression, and controls and patients with self-reported depression were drawn from QSkin, a population-based cohort study. Data analyzed herein were collected before September 2018, and data analysis was conducted from September 10, 2020, to January 27, 2021. Main Outcome and Measures: Polygenic risk scores generated from genome-wide association studies using different definitions of depression were evaluated for estimation of MDD in and within individuals with MDD for an association with age at onset, adverse childhood experiences, comorbid psychiatric and somatic disorders, and current physical and mental health. Results: Participants included 12 106 (71% female; mean age, 42.3 years; range, 18-88 years) patients meeting criteria for MDD and 12 621 (55% female; mean age, 60.9 years; range, 43-87 years) control participants with no history of psychiatric disorders. The effect size of the PRS was proportional to the discovery sample size, with the largest study having the largest effect size with the odds ratio for MDD (1.75; 95% CI, 1.73-1.77) per SD of PRS and the PRS derived from ICD-10 codes documented in hospitalization records in a population health cohort having the lowest odds ratio (1.14; 95% CI, 1.12-1.16). When accounting for differences in sample size, the PRS from a genome-wide association study of patients meeting diagnostic criteria for MDD and control participants was the best estimator of MDD, but not in those with self-reported depression, and associations with higher odds ratios with childhood adverse experiences and measures of somatic distress. Conclusions and Relevance: These findings suggest that increasing sample sizes, regardless of the depth of phenotyping, may be most informative for estimating risk of depression. The next generation of genome-wide association studies should, like the Australian Genetics of Depression Study, have both large sample sizes and extensive phenotyping to capture genetic risk factors for MDD not identified by other definitions of depression.


Subject(s)
Depressive Disorder, Major/genetics , Genome-Wide Association Study , Multifactorial Inheritance/genetics , Adult , Aged , Case-Control Studies , Depressive Disorder, Major/physiopathology , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Phenotype , Risk , Sample Size
14.
Nat Hum Behav ; 5(10): 1432-1442, 2021 10.
Article in English | MEDLINE | ID: mdl-33859377

ABSTRACT

Depression and anxiety are highly prevalent and comorbid psychiatric traits that cause considerable burden worldwide. Here we use factor analysis and genomic structural equation modelling to investigate the genetic factor structure underlying 28 items assessing depression, anxiety and neuroticism, a closely related personality trait. Symptoms of depression and anxiety loaded on two distinct, although highly genetically correlated factors, and neuroticism items were partitioned between them. We used this factor structure to conduct genome-wide association analyses on latent factors of depressive symptoms (89 independent variants, 61 genomic loci) and anxiety symptoms (102 variants, 73 loci) in the UK Biobank. Of these associated variants, 72% and 78%, respectively, replicated in an independent cohort of approximately 1.9 million individuals with self-reported diagnosis of depression and anxiety. We use these results to characterize shared and trait-specific genetic associations. Our findings provide insight into the genetic architecture of depression and anxiety and comorbidity between them.


Subject(s)
Anxiety , Behavioral Symptoms , Depression , Neuroticism/physiology , Anxiety/diagnosis , Anxiety/epidemiology , Anxiety/genetics , Behavioral Symptoms/diagnosis , Behavioral Symptoms/psychology , Comorbidity , Depression/diagnosis , Depression/epidemiology , Depression/genetics , Factor Analysis, Statistical , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Latent Class Analysis , Symptom Assessment/methods , Symptom Assessment/statistics & numerical data
15.
Commun Med (Lond) ; 1: 45, 2021.
Article in English | MEDLINE | ID: mdl-35602235

ABSTRACT

Background: Major depression is one of the most disabling health conditions internationally. In recent years, new generation antidepressant medicines have become very widely prescribed. While these medicines are efficacious, side effects are common and frequently result in discontinuation of treatment. Compared with specific pharmacological properties of the different medications, the relevance of individual vulnerability is understudied. Methods: We used data from the Australian Genetics of Depression Study to gain insights into the aetiology and genetic risk factors to antidepressant side effects. To this end, we employed structural equation modelling, polygenic risk scoring and regressions. Results: Here we show that participants reporting a specific side effect for one antidepressant are more likely to report the same side effect for other antidepressants, suggesting the presence of shared individual or pharmacological factors. Polygenic risk scores (PRS) for depression associated with side effects that overlapped with depressive symptoms, including suicidality and anxiety. Body Mass Index PRS are strongly associated with weight gain from all medications. PRS for headaches are associated with headaches from sertraline. Insomnia PRS show some evidence of predicting insomnia from amitriptyline and escitalopram. Conclusions: Our results suggest a set of common factors underlying the risk for antidepressant side effects. These factors seem to be partly explained by genetic liability related to depression severity and the nature of the side effect. Future studies on the genetic aetiology of side effects will enable insights into their underlying mechanisms and the possibility of risk stratification and prophylaxis strategies.

17.
Alzheimers Dement (Amst) ; 12(1): e12108, 2020.
Article in English | MEDLINE | ID: mdl-33005726

ABSTRACT

INTRODUCTION: Hearing loss has been identified as the potentially largest modifiable risk factor for Alzheimer's disease (AD), estimated to account for a similar increase in AD risk as the apolipoprotein E (APOE) gene. METHODS: We investigated the genetic relationship between hearing loss and AD, and sought evidence for a causal relationship. RESULTS: We found a significant genetic overlap between hearing impairment and AD and a polygenic risk score for AD was able to significantly predict hearing loss in an independent cohort. Additionally, regions of the genome involved in inflammation were identified to be shared between hearing difficulty and AD. However, causality tests found no significant evidence of a causal relationship between these traits in either direction. DISCUSSION: Overall, these results show that the relationship between hearing difficulty and AD may, in part, be due to shared genes and immune response pathways between the traits. However, currently available data do not support a causal relationship.

18.
Twin Res Hum Genet ; 23(4): 221-227, 2020 08.
Article in English | MEDLINE | ID: mdl-32885772

ABSTRACT

There is a well-established relationship between cannabis use and psychosis, although the exact nature of this relationship is not fully understood. Recent studies have observed significant genetic overlap between a diagnosis of schizophrenia and lifetime cannabis use. Expanding on this work, the current study aimed to examine whether genetic overlap also occurs for subclinical psychosis (schizotypy) and cannabis use, as well as examining the phenotypic association between the traits. Phenotypic correlations were calculated for a variety of schizotypy and cannabis phenotypes in the UK Biobank (UKB), and single nucleotide polymorphism (SNP)-based heritability estimates and genetic correlations were calculated for these UKB phenotypes as well as for several other variables taken from recent genomewide association studies. Positive phenotypic correlations were observed between 11 out of 12 pairs of the cannabis use and schizotypy phenotypes (correlation range .05-.18), indicating a robust association between increased symptoms of schizotypy and cannabis use. SNP-based heritability estimates for two schizotypy phenotypes remained significant after multiple testing correction: social anhedonia (h2SNP = .08, SE = .02, N = 4025) and ever seen an unreal vision (h2SNP = .35, SE = .10, N = 150,717). Finally, one significant genetic correlation was observed between schizotypy and cannabis use, a negative correlation between social anhedonia and number of times used cannabis (rg = -.30, p = .012). The current study suggests the relationship between cannabis use and psychosis is also seen in subclinical symptoms of psychosis, but further research with larger samples is needed to determine the biological mechanisms underlying this association.


Subject(s)
Marijuana Use/genetics , Psychotic Disorders , Schizotypal Personality Disorder , Cannabis , Humans , Phenotype , Polymorphism, Single Nucleotide , Psychotic Disorders/genetics , Schizotypal Personality Disorder/genetics
19.
Psychol Med ; 50(14): 2385-2396, 2020 10.
Article in English | MEDLINE | ID: mdl-31530331

ABSTRACT

BACKGROUND: Depression is a clinically heterogeneous disorder. Previous large-scale genetic studies of depression have explored genetic risk factors of depression case-control status or aggregated sums of depressive symptoms, ignoring possible clinical or genetic heterogeneity. METHODS: We analyse data from 148 752 subjects of white British ancestry in the UK Biobank who completed nine items of a self-rated measure of current depressive symptoms: the Patient Health Questionnaire (PHQ-9). Genome-Wide Association analyses were conducted for nine symptoms and two composite measures. LD Score Regression was used to calculate SNP-based heritability (h2SNP) and genetic correlations (rg) across symptoms and to investigate genetic correlations with 25 external phenotypes. Genomic structural equation modelling was used to test the genetic factor structure across the nine symptoms. RESULTS: We identified nine genome-wide significant genomic loci (8 novel), with no overlap in loci across symptoms. h2SNP ranged from 6% (concentration problems) to 9% (appetite changes). Genetic correlations ranged from 0.54 to 0.96 (all p < 1.39 × 10-3) with 30 of 36 correlations being significantly smaller than one. A two-factor model provided the best fit to the genetic covariance matrix, with factors representing 'psychological' and 'somatic' symptoms. The genetic correlations with external phenotypes showed large variation across the nine symptoms. CONCLUSIONS: Patterns of SNP associations and genetic correlations differ across the nine symptoms, suggesting that current depressive symptoms are genetically heterogeneous. Our study highlights the value of symptom-level analyses in understanding the genetic architecture of a psychiatric trait. Future studies should investigate whether genetic heterogeneity is recapitulated in clinical symptoms of major depression.


Subject(s)
Depression/genetics , Genetic Heterogeneity , Genetic Loci , Genetic Predisposition to Disease , Aged , Aged, 80 and over , Case-Control Studies , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Patient Health Questionnaire , Phenotype , Self Report , United Kingdom , White People/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...