Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
medRxiv ; 2024 May 25.
Article in English | MEDLINE | ID: mdl-38826220

ABSTRACT

The brain's default mode network (DMN) plays a role in social cognition, with altered DMN function being associated with social impairments across various neuropsychiatric disorders. In the present study, we examined the genetic relationship between sociability and DMN-related resting-state functional magnetic resonance imaging (rs-fMRI) traits. To this end, we used genome-wide association summary statistics for sociability and 31 activity and 64 connectivity DMN-related rs-fMRI traits (N=34,691-342,461). First, we examined global and local genetic correlations between sociability and the rs-fMRI traits. Second, to assess putatively causal relationships between the traits, we conducted bi-directional Mendelian randomisation (MR) analyses. Finally, we prioritised genes influencing both sociability and rs-fMRI traits by combining three methods: gene-expression eQTL MR analyses, the CELLECT framework using single-nucleus RNA-seq data, and network propagation in the context of a protein-protein interaction network. Significant local genetic correlations were found between sociability and two rs-fMRI traits, one representing spontaneous activity within the temporal cortex, the other representing connectivity between the frontal/cingulate and angular/temporal cortices. Sociability affected 12 rs-fMRI traits when allowing for weakly correlated genetic instruments. Combing all three methods for gene prioritisation, we defined 17 highly prioritised genes, with DRD2 and LINGO1 showing the most robust evidence across all analyses. By integrating genetic and transcriptomics data, our gene prioritisation strategy may serve as a blueprint for future studies. The prioritised genes could be explored as potential biomarkers for social dysfunction in the context of neuropsychiatric disorders and as drug target genes.

2.
medRxiv ; 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38496672

ABSTRACT

The co-occurrence of insulin resistance (IR)-related metabolic conditions with neuropsychiatric disorders is a complex public health challenge. Evidence of the genetic links between these phenotypes is emerging, but little is currently known about the genomic regions and biological functions that are involved. To address this, we performed Local Analysis of [co]Variant Association (LAVA) using large-scale (N=9,725-933,970) genome-wide association studies (GWASs) results for three IR-related conditions (type 2 diabetes mellitus, obesity, and metabolic syndrome) and nine neuropsychiatric disorders. Subsequently, positional and expression quantitative trait locus (eQTL)-based gene mapping and downstream functional genomic analyses were performed on the significant loci. Patterns of negative and positive local genetic correlations (|rg|=0.21-1, pFDR<0.05) were identified at 109 unique genomic regions across all phenotype pairs. Local correlations emerged even in the absence of global genetic correlations between IR-related conditions and Alzheimer's disease, bipolar disorder, and Tourette's syndrome. Genes mapped to the correlated regions showed enrichment in biological pathways integral to immune-inflammatory function, vesicle trafficking, insulin signalling, oxygen transport, and lipid metabolism. Colocalisation analyses further prioritised 10 genetically correlated regions for likely harbouring shared causal variants, displaying high deleterious or regulatory potential. These variants were found within or in close proximity to genes, such as SLC39A8 and HLA-DRB1, that can be targeted by supplements and already known drugs, including omega-3/6 fatty acids, immunomodulatory, antihypertensive, and cholesterol-lowering drugs. Overall, our findings underscore the complex genetic landscape of IR-neuropsychiatric multimorbidity, advocating for an integrated disease model and offering novel insights for research and treatment strategies in this domain.

3.
Am J Med Genet B Neuropsychiatr Genet ; 195(1): e32951, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37334623

ABSTRACT

The dense co-occurrence of psychiatric disorders questions the categorical classification tradition and motivates efforts to establish dimensional constructs with neurobiological foundations that transcend diagnostic boundaries. In this study, we examined the genetic liability for eight major psychiatric disorder phenotypes under both a disorder-specific and a transdiagnostic framework. The study sample (n = 513) was deeply phenotyped, consisting of 452 patients from tertiary care with mood disorders, anxiety disorders (ANX), attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorders, and/or substance use disorders (SUD) and 61 unaffected comparison individuals. We computed subject-specific polygenic risk score (PRS) profiles and assessed their associations with psychiatric diagnoses, comorbidity status, as well as cross-disorder behavioral dimensions derived from a rich battery of psychopathology assessments. High PRSs for depression were unselectively associated with the diagnosis of SUD, ADHD, ANX, and mood disorders (p < 1e-4). In the dimensional approach, four distinct functional domains were uncovered, namely the negative valence, social, cognitive, and regulatory systems, closely matching the major functional domains proposed by the Research Domain Criteria (RDoC) framework. Critically, the genetic predisposition for depression was selectively reflected in the functional aspect of negative valence systems (R2 = 0.041, p = 5e-4) but not others. This study adds evidence to the ongoing discussion about the misalignment between current psychiatric nosology and the underlying psychiatric genetic etiology and underscores the effectiveness of the dimensional approach in both the functional characterization of psychiatric patients and the delineation of the genetic liability for psychiatric disorders.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Psychiatry , Substance-Related Disorders , Humans , Attention Deficit Disorder with Hyperactivity/diagnosis , Attention Deficit Disorder with Hyperactivity/genetics , Psychopathology , Anxiety Disorders , Multifactorial Inheritance/genetics
4.
medRxiv ; 2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36711661

ABSTRACT

Importance: Psychiatric disorders can have an immense impact on socioeconomic, physical, and social-psychological facets of life. Psychiatric disorders are also highly heritable. Under a liability threshold model, an important question arises as to what extent genetic liability for psychiatric disorders relates to, and possibly impacts on, different aspects of quality of life in the general population. Objective: To characterize the link between psychiatric genetic liability and diverse aspects of quality of life in childhood and adulthood. Design setting and participants: We used data from two multi-site, population-based cohorts, i.e. preadolescent children in the USA enrolled at age 9-10 years from the Adolescent Brain Cognitive Development (ABCD) study (N=4,645) and white British adults between age 40-69 years from the UK Biobank (UKB) study (N=377,664). Due to the current limitations of our genetic methods, only data from unrelated individuals of European descent could be included. Main outcomes and measures: To derive robust measures capturing multiple domains of quality of life in each of the cohorts, we integrated an array of measurements of academic, economic, and physical status, as well as social well-being, in a second-level three-factor confirmatory factor analysis. The genetic liabilities to seven major psychiatric disorders were quantified by a set of polygenic scores (PGSs) derived from the largest genome-wide association studies to date, independent of the target cohorts, of major depressive disorder (MDD, N=142k-173k), anxiety disorders (ANX, N=22k-144k), attention-deficit/hyperactivity disorder (ADHD, N=226k), autism spectrum disorder (ASD, N=55k), schizophrenia (SCZ, N=130k), bipolar disorder (BIP, N=353k-414k), and cannabis use disorder (CUD, N=384k). Using general linear models we assessed associations between PGSs and the estimated latent factors, controlling for age, sex, site, genotyping batch, plate, and genetic ancestry. Results: In each cohort, three latent factors indexing distinct but correlated quality of life domains, (1) educational performance and cognition (Edu, in ABCD) / social economic status (SES, in UKB), (2) physical health (Hea), (3) adverse social experience (Adv, in ABCD) / social well-being (Soc, in UKB), were estimated with excellent model fit indices. In addition, a general factor was derived that captured the covariances between the three latent factors (QoL). In the ABCD cohort, ADHD-PGS was significantly associated with Edu (ß = -0.13, t = -8.29, p = 1.53e-16), Adv (ß = -0.09, t = -5.79, p = 7.81e-09), and general QoL (ß = -0.14, t = -8.74, p = 3.37e-18) factors. In the UKB cohort, all examined disorder PGSs were significantly associated with the general QoL latent factor and at least one first-order subdomain, with ADHD-PGS (ß = -0.06 ~ -0.10, t = -29.1 ~ -52.5, p < 5.91e-186) and MDD-PGS (ß = -0.04 ~ -0.07, t = -23.8 ~ -36.3, p < 3.63e-125) showing the largest effects. Conclusions and relevance: The present study reveals an inverse relationship between psychiatric genetic liabilities and multiple quality of life metrics, with ADHD-associated genetic risk being the main contributor in both children and adults, and MDD additionally showing effects in adults. All effect sizes observed were small, as expected. Understanding potential real-world outcomes of quantitative measures of disorder-related genetic risks in the general population can provide a scientific foundation for societal intervention and policy-making processes, with profound implications for promoting a flourishing society.

5.
Neurosci Biobehav Rev ; 143: 104927, 2022 12.
Article in English | MEDLINE | ID: mdl-36367493

ABSTRACT

Clinical and genomic studies have shown an overlap between neuropsychiatric disorders and insulin resistance (IR)-related somatic conditions, including obesity, type 2 diabetes, and cardiovascular diseases. Impaired cognition is often observed among neuropsychiatric disorders, where multiple cognitive domains may be affected. In this review, we aimed to summarise previous evidence on the relationship between IR-related diseases/traits and cognitive performance in the large UK Biobank study cohort. Electronic searches were conducted on PubMed, Scopus, and Web of Science until April 2022. Eighteen articles met the inclusion criteria and were qualitatively reviewed. Overall, there is substantial evidence for an association between IR-related cardio-metabolic diseases/traits and worse performance on various cognitive domains, which is largely independent of possible confoundings. The most consistent findings referred to IR-related associations with poorer verbal and numerical reasoning ability, as well as slower processing speed. The observed associations might be mediated by alterations in immune-inflammation, brain integrity/connectivity, and/or comorbid somatic or psychiatric diseases/traits. Our findings provide impetus for further research into the underlying neurobiology and possible new therapeutic targets.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Biological Specimen Banks , Cognition , United Kingdom/epidemiology
6.
Mol Psychiatry ; 27(11): 4453-4463, 2022 11.
Article in English | MEDLINE | ID: mdl-36284158

ABSTRACT

Despite the substantial heritability of antisocial behavior (ASB), specific genetic variants robustly associated with the trait have not been identified. The present study by the Broad Antisocial Behavior Consortium (BroadABC) meta-analyzed data from 28 discovery samples (N = 85,359) and five independent replication samples (N = 8058) with genotypic data and broad measures of ASB. We identified the first significant genetic associations with broad ASB, involving common intronic variants in the forkhead box protein P2 (FOXP2) gene (lead SNP rs12536335, p = 6.32 × 10-10). Furthermore, we observed intronic variation in Foxp2 and one of its targets (Cntnap2) distinguishing a mouse model of pathological aggression (BALB/cJ strain) from controls (BALB/cByJ strain). Polygenic risk score (PRS) analyses in independent samples revealed that the genetic risk for ASB was associated with several antisocial outcomes across the lifespan, including diagnosis of conduct disorder, official criminal convictions, and trajectories of antisocial development. We found substantial genetic correlations of ASB with mental health (depression rg = 0.63, insomnia rg = 0.47), physical health (overweight rg = 0.19, waist-to-hip ratio rg = 0.32), smoking (rg = 0.54), cognitive ability (intelligence rg = -0.40), educational attainment (years of schooling rg = -0.46) and reproductive traits (age at first birth rg = -0.58, father's age at death rg = -0.54). Our findings provide a starting point toward identifying critical biosocial risk mechanisms for the development of ASB.


Subject(s)
Antisocial Personality Disorder , Conduct Disorder , Animals , Mice , Antisocial Personality Disorder/genetics , Genome-Wide Association Study , Conduct Disorder/genetics , Conduct Disorder/psychology , Aggression/psychology , Multifactorial Inheritance/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics
7.
Mol Psychiatry ; 27(11): 4464-4473, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35948661

ABSTRACT

Common variation in the gene encoding the neuron-specific RNA splicing factor RNA Binding Fox-1 Homolog 1 (RBFOX1) has been identified as a risk factor for several psychiatric conditions, and rare genetic variants have been found causal for autism spectrum disorder (ASD). Here, we explored the genetic landscape of RBFOX1 more deeply, integrating evidence from existing and new human studies as well as studies in Rbfox1 knockout mice. Mining existing data from large-scale studies of human common genetic variants, we confirmed gene-based and genome-wide association of RBFOX1 with risk tolerance, major depressive disorder and schizophrenia. Data on six mental disorders revealed copy number losses and gains to be more frequent in ASD cases than in controls. Consistently, RBFOX1 expression appeared decreased in post-mortem frontal and temporal cortices of individuals with ASD and prefrontal cortex of individuals with schizophrenia. Brain-functional MRI studies demonstrated that carriers of a common RBFOX1 variant, rs6500744, displayed increased neural reactivity to emotional stimuli, reduced prefrontal processing during cognitive control, and enhanced fear expression after fear conditioning, going along with increased avoidance behaviour. Investigating Rbfox1 neuron-specific knockout mice allowed us to further specify the role of this gene in behaviour. The model was characterised by pronounced hyperactivity, stereotyped behaviour, impairments in fear acquisition and extinction, reduced social interest, and lack of aggression; it provides excellent construct and face validity as an animal model of ASD. In conclusion, convergent translational evidence shows that common variants in RBFOX1 are associated with a broad spectrum of psychiatric traits and disorders, while rare genetic variation seems to expose to early-onset neurodevelopmental psychiatric disorders with and without developmental delay like ASD, in particular. Studying the pleiotropic nature of RBFOX1 can profoundly enhance our understanding of mental disorder vulnerability.


Subject(s)
Autism Spectrum Disorder , Depressive Disorder, Major , Mental Disorders , Animals , Mice , Humans , Autism Spectrum Disorder/genetics , Depressive Disorder, Major/genetics , Genome-Wide Association Study , Mental Disorders/genetics , Mice, Knockout , RNA Splicing Factors/genetics
8.
Neurosci Biobehav Rev ; 139: 104757, 2022 08.
Article in English | MEDLINE | ID: mdl-35777579

ABSTRACT

ADHD is a major burden in adulthood, where co-morbid conditions such as depression, substance use disorder and obesity often dominate the clinical picture. ADHD has substantial shared heritability with other mental disorders, contributing to comorbidity. However, environmental risk factors exist but their interaction with genetic makeup, especially in relation to comorbid disorders, remains elusive. This review for the first time summarizes present knowledge on gene x environment (GxE) interactions regarding the dopamine system. Hitherto, mainly candidate (GxE) studies were performed, focusing on the genes DRD4, DAT1 and MAOA. Some evidence suggest that the variable number tandem repeats in DRD4 and MAOA may mediate GxE interactions in ADHD generally, and comorbid conditions specifically. Nevertheless, even for these genes, common variants are bound to suggest risk only in the context of gender and specific environments. For other polymorphisms, evidence is contradictory and less convincing. Particularly lacking are longitudinal studies testing the interaction of well-defined environmental factors with polygenic risk scores reflecting the dopamine system in its entirety.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Receptors, Dopamine D4 , Adult , Attention Deficit Disorder with Hyperactivity/epidemiology , Attention Deficit Disorder with Hyperactivity/genetics , Comorbidity , Dopamine/genetics , Dopamine Plasma Membrane Transport Proteins/genetics , Genetic Predisposition to Disease/genetics , Humans , Morbidity , Receptors, Dopamine D4/genetics
9.
Int J Mol Sci ; 23(6)2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35328598

ABSTRACT

Externalizing behavior in its more extreme form is often considered a problem to the individual, their families, teachers, and society as a whole. Several brain structures have been linked to externalizing behavior and such associations may arise if the (co)development of externalizing behavior and brain structures share the same genetic and/or environmental factor(s). We assessed externalizing behavior with the Child Behavior Checklist and Youth Self Report, and the brain volumes and white matter integrity (fractional anisotropy [FA] and mean diffusivity [MD]) with magnetic resonance imaging in the BrainSCALE cohort, which consisted of twins and their older siblings from 112 families measured longitudinally at ages 10, 13, and 18 years for the twins. Genetic covariance modeling based on the classical twin design, extended to also include siblings of twins, showed that genes influence externalizing behavior and changes therein (h2 up to 88%). More pronounced externalizing behavior was associated with higher FA (observed correlation rph up to +0.20) and lower MD (rph up to -0.20), with sizeable genetic correlations (FA ra up to +0.42; MD ra up to -0.33). The cortical gray matter (CGM; rph up to -0.20) and cerebral white matter (CWM; rph up to +0.20) volume were phenotypically but not genetically associated with externalizing behavior. These results suggest a potential mediating role for global brain structures in the display of externalizing behavior during adolescence that are both partially explained by the influence of the same genetic factor.


Subject(s)
Twins , White Matter , Adolescent , Brain/diagnostic imaging , Child , Genetic Structures , Humans , Magnetic Resonance Imaging , Twins/genetics , White Matter/diagnostic imaging
10.
Transl Psychiatry ; 12(1): 59, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35165256

ABSTRACT

The prevalence of somatic insulinopathies, like metabolic syndrome (MetS), obesity, and type 2 diabetes mellitus (T2DM), is higher in Alzheimer's disease (AD), autism spectrum disorder (ASD), and obsessive-compulsive disorder (OCD). Dysregulation of insulin signalling has been implicated in these neuropsychiatric disorders, and shared genetic factors might partly underlie this observed multimorbidity. We investigated the genetic overlap between AD, ASD, and OCD with MetS, obesity, and T2DM by estimating pairwise global genetic correlations using the summary statistics of the largest available genome-wide association studies for these phenotypes. Having tested these hypotheses, other potential brain "insulinopathies" were also explored by estimating the genetic relationship of six additional neuropsychiatric disorders with nine insulin-related diseases/traits. Stratified covariance analyses were then performed to investigate the contribution of insulin-related gene sets. Significant negative genetic correlations were found between OCD and MetS (rg = -0.315, p = 3.9 × 10-8), OCD and obesity (rg = -0.379, p = 3.4 × 10-5), and OCD and T2DM (rg = -0.172, p = 3 × 10-4). Significant genetic correlations with insulin-related phenotypes were also found for anorexia nervosa (AN), attention-deficit/hyperactivity disorder (ADHD), major depressive disorder, and schizophrenia (p < 6.17 × 10-4). Stratified analyses showed negative genetic covariances between AD, ASD, OCD, ADHD, AN, bipolar disorder, schizophrenia and somatic insulinopathies through gene sets related to insulin signalling and insulin receptor recycling, and positive genetic covariances between AN and T2DM, as well as ADHD and MetS through gene sets related to insulin processing/secretion (p < 2.06 × 10-4). Overall, our findings suggest the existence of two clusters of neuropsychiatric disorders, in which the genetics of insulin-related diseases/traits may exert divergent pleiotropic effects. These results represent a starting point for a new research line on "insulinopathies" of the brain.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Autism Spectrum Disorder , Depressive Disorder, Major , Diabetes Mellitus, Type 2 , Attention Deficit Disorder with Hyperactivity/psychology , Autism Spectrum Disorder/psychology , Brain , Depressive Disorder, Major/genetics , Diabetes Mellitus, Type 2/genetics , Genome-Wide Association Study , Humans , Insulin
11.
Schizophr Bull ; 48(3): 551-562, 2022 05 07.
Article in English | MEDLINE | ID: mdl-35137221

ABSTRACT

INTRODUCTION: Illuminating neurobiological mechanisms underlying the protective effect of recently discovered common genetic resilience variants for schizophrenia is crucial for more effective prevention efforts. Current models implicate adaptive neuroplastic changes in the visual system and their pro-cognitive effects as a schizophrenia resilience mechanism. We investigated whether common genetic resilience variants might affect brain structure in similar neural circuits. METHOD: Using structural magnetic resonance imaging, we measured the impact of an established schizophrenia polygenic resilience score (PRSResilience) on cortical volume, thickness, and surface area in 101 healthy subjects and in a replication sample of 33 224 healthy subjects (UK Biobank). FINDING: We observed a significant positive whole-brain correlation between PRSResilience and cortical volume in the right fusiform gyrus (FFG) (r = 0.35; P = .0004). Post-hoc analyses in this cluster revealed an impact of PRSResilience on cortical surface area. The replication sample showed a positive correlation between PRSResilience and global cortical volume and surface area in the left FFG. CONCLUSION: Our findings represent the first evidence of a neurobiological correlate of a genetic resilience factor for schizophrenia. They support the view that schizophrenia resilience emerges from strengthening neural circuits in the ventral visual pathway and an increased capacity for the disambiguation of social and nonsocial visual information. This may aid psychosocial functioning, ameliorate the detrimental effects of subtle perceptual and cognitive disturbances in at-risk individuals, and facilitate coping with the cognitive and psychosocial consequences of stressors. Our results thus provide a novel link between visual cognition, the vulnerability-stress concept, and schizophrenia resilience models.


Subject(s)
Schizophrenia , Brain/metabolism , Humans , Magnetic Resonance Imaging , Multifactorial Inheritance , Schizophrenia/diagnostic imaging , Schizophrenia/genetics , Schizophrenia/metabolism , Visual Pathways/diagnostic imaging , Visual Pathways/pathology
12.
Psychol Med ; 51(13): 2274-2286, 2021 10.
Article in English | MEDLINE | ID: mdl-33814023

ABSTRACT

Attention-deficit/hyperactivity disorder (ADHD) is a common and highly heritable neurodevelopmental disorder (NDD). In this narrative review, we summarize recent advances in quantitative and molecular genetic research from the past 5-10 years. Combined with large-scale international collaboration, these advances have resulted in fast-paced progress in understanding the etiology of ADHD and how genetic risk factors map on to clinical heterogeneity. Studies are converging on a number of key insights. First, ADHD is a highly polygenic NDD with a complex genetic architecture encompassing risk variants across the spectrum of allelic frequencies, which are implicated in neurobiological processes. Second, genetic studies strongly suggest that ADHD diagnosis shares a large proportion of genetic risks with continuously distributed traits of ADHD in the population, with shared genetic risks also seen across development and sex. Third, ADHD genetic risks are shared with those implicated in many other neurodevelopmental, psychiatric and somatic phenotypes. As sample sizes and the diversity of genetic studies continue to increase through international collaborative efforts, we anticipate further success with gene discovery, characterization of how the ADHD phenotype relates to other human traits and growing potential to use genomic risk factors for understanding clinical trajectories and for precision medicine approaches.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Genome-Wide Association Study , Multifactorial Inheritance/genetics , Attention Deficit Disorder with Hyperactivity/epidemiology , Attention Deficit Disorder with Hyperactivity/genetics , Humans , Molecular Biology , Phenotype , Risk Factors
13.
Am J Med Genet B Neuropsychiatr Genet ; 186(7): 401-411, 2021 10.
Article in English | MEDLINE | ID: mdl-32815639

ABSTRACT

This study investigated the genetic components of ADHD and ASD by examining the cross-disorder trait of emotion recognition problems. The genetic burden for ADHD and ASD on previously identified emotion recognition factors (speed and accuracy of visual and auditory emotion recognition) and classes (Class 1: Average visual, impulsive auditory; Class 2: Average-strong visual & auditory; Class 3: Impulsive & imprecise visual, average auditory; Class 4: Weak visual & auditory) was assessed using ASD and ADHD polygenic risk scores (PRS). Our sample contained 552 participants: 74 with ADHD, 85 with ASD, 60 with ASD + ADHD, 177 unaffected siblings of ADHD or ASD probands, and 156 controls. ADHD- and ASD-PRS, calculated from the latest ADHD and ASD GWAS meta-analyses, were analyzed across these emotion recognition factors and classes using linear mixed models. Unexpectedly, the analysis of emotion recognition factors showed higher ASD-PRS to be associated with faster visual emotion recognition. The categorical analysis of emotion recognition classes showed ASD-PRS to be reduced in Class 3 compared to the other classes (p value threshold [pT] = 1, p = .021). A dimensional analysis identified a high ADHD-PRS reduced the probability of being assigned to the Class 1 or Class 3 (pT = .05, p = .028 and p = .044, respectively). Though these nominally significant results did not pass FDR correction, they potentially indicate different indirect causative chains from genetics via emotion recognition to ADHD and ASD, which need to be verified in future research.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Autism Spectrum Disorder , Attention Deficit Disorder with Hyperactivity/genetics , Autism Spectrum Disorder/genetics , Emotions , Humans , Risk Factors , Siblings
14.
J Psychiatr Res ; 136: 529-536, 2021 04.
Article in English | MEDLINE | ID: mdl-33127071

ABSTRACT

The association between obesity and attention-deficit hyperactivity disorder (ADHD) has been extensively reported in the literature. However, the potential mechanisms underlying this association are not completely understood. This study aimed to evaluate the association between body composition and ADHD and explore the possible genetic mechanisms involved. We used data from the 1982 Pelotas (Brazil) Birth Cohort at age 30-year follow-up (N = 3630). We first used logistic regression analysis to test whether body mass index (BMI), fat mass (FM), and fat-free mass (FFM) were associated with ADHD. We further tested the association between BMI polygenic risk score (BMI-PRS) and ADHD and the role of the genes upregulated in the reward system using a gene-set association approach. BMI (odds ratio [OR] = 1.05; 95% confidence interval [CI], 1.00-1.09; p = 0.038) and FM (OR = 1.04; 95% CI, 1.00-1.07; p = 0.043) were associated with ADHD. The BMI-PRS was associated with ADHD (using p-value threshold (PT) = 0.4; OR = 1.65; 95% CI, 1.02-2.65) at a nominal level. In gene-set analysis, the reward system genes were associated with BMI in subjects with a high BMI-PRS score, considering PT = 0.4 (p = 0.014). The results suggest that BMI genetic components, especially those genes related to the reward system, may be involved in this association.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Adult , Attention Deficit Disorder with Hyperactivity/genetics , Body Composition , Body Mass Index , Brazil , Humans , Obesity/epidemiology , Obesity/genetics , Reward , Risk Factors
15.
Am J Med Genet B Neuropsychiatr Genet ; 186(4): 242-250, 2021 06.
Article in English | MEDLINE | ID: mdl-33319511

ABSTRACT

Attention-deficit/hyperactivity disorder (ADHD) symptoms are continuously distributed in the general population, where both genetic and environmental factors play roles. Stressful life events (SLEs) have been associated with ADHD diagnosis, but the relationship between ADHD genetic liability, SLEs, and ADHD symptoms in healthy individuals is less clear. Using a sample of 1,531 healthy adults (average age 26.9 years; 55.8% female), we investigated relationships between ADHD polygenic risk scores (ADHD-PRSs), SLEs, and ADHD symptoms in a general population sample. Confirming earlier findings in an overlapping sample, all SLE-measures assessed (lifetime SLEs, recent SLEs, and childhood trauma (CT)) were significantly correlated with total ADHD, inattention (IA), and hyperactivity-impulsivity (HI) scores (r2 range = .08-.15; all p < .005). ADHD-PRSs was associated with HI (R2best-fit  = .37%), lifetime SLEs (R2best-fit  = .56%), and CT (R2best-fit  = .40%). Mediation analyses showed that lifetime SLEs partially mediated the association between ADHD-PRSs and HI (indirect effect: ß = 68.6, bias corrected accelerated 95% confident interval (BCa95%CI) [11.9, 131.0], p = .016, proportion mediated (PM ) =19.5%), with strongest effects contributed by CT (ß = 34.4, BCa95%CI [0.4, 76.5], p = .040, PM  = 9.8%). On the other hand, HI partially mediated the association between the ADHD-PRSs and lifetime SLEs (ß = 42.9, BCa95%CI [7.3, 83.9], p = .014, PM  = 18.8%). Our study observed a complex relationship of genetic and environmental risk factors contributing to ADHD symptoms in the healthy adult population.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Stress, Psychological/genetics , Adult , Chromosome Mapping/methods , Cognition , Female , Genetic Association Studies/methods , Genetic Predisposition to Disease/genetics , Healthy Volunteers , Humans , Life Change Events , Longitudinal Studies , Male , Phenotype , Stress, Psychological/metabolism
16.
Neuropsychopharmacology ; 45(10): 1617-1626, 2020 09.
Article in English | MEDLINE | ID: mdl-32279069

ABSTRACT

Attention deficit/hyperactivity disorder (ADHD) is a common neurodevelopmental disorder characterized by age-inappropriate symptoms of inattention, impulsivity, and hyperactivity that persist into adulthood in the majority of the diagnosed children. Despite several risk factors during childhood predicting the persistence of ADHD symptoms into adulthood, the genetic architecture underlying the trajectory of ADHD over time is still unclear. We set out to study the contribution of common genetic variants to the risk for ADHD across the lifespan by conducting meta-analyses of genome-wide association studies on persistent ADHD in adults and ADHD in childhood separately and jointly, and by comparing the genetic background between them in a total sample of 17,149 cases and 32,411 controls. Our results show nine new independent loci and support a shared contribution of common genetic variants to ADHD in children and adults. No subgroup heterogeneity was observed among children, while this group consists of future remitting and persistent individuals. We report similar patterns of genetic correlation of ADHD with other ADHD-related datasets and different traits and disorders among adults, children, and when combining both groups. These findings confirm that persistent ADHD in adults is a neurodevelopmental disorder and extend the existing hypothesis of a shared genetic architecture underlying ADHD and different traits to a lifespan perspective.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Adult , Attention Deficit Disorder with Hyperactivity/genetics , Child , Genetic Background , Genome-Wide Association Study , Humans , Impulsive Behavior , Phenotype
17.
Am J Psychiatry ; 177(6): 526-536, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32046534

ABSTRACT

OBJECTIVE: Attention deficit hyperactivity disorder (ADHD) is a common, highly heritable neuropsychiatric disorder. ADHD often co-occurs with intellectual disability, and shared overlapping genetics have been suggested. The aim of this study was to identify novel ADHD genes by investigating whether genes carrying rare mutations linked to intellectual disability contribute to ADHD risk through common genetic variants. Validation and characterization of candidates were performed using Drosophila melanogaster. METHODS: Common genetic variants in a diagnostic gene panel of 396 autosomal intellectual disability genes were tested for association with ADHD risk through gene set and gene-wide analyses, using ADHD meta-analytic data from the Psychiatric Genomics Consortium for discovery (N=19,210) and ADHD data from the Lundbeck Foundation Initiative for Integrative Psychiatric Research for replication (N=37,076). The significant genes were functionally validated and characterized in Drosophila by assessing locomotor activity and sleep upon knockdown of those genes in brain circuits. RESULTS: The intellectual disability gene set was significantly associated with ADHD risk in the discovery and replication data sets. The three genes most consistently associated were MEF2C, ST3GAL3, and TRAPPC9. Performing functional characterization of the two evolutionarily conserved genes in Drosophila melanogaster, the authors found that their knockdown in dopaminergic (dMEF2) and circadian neurons (dTRAPPC9) resulted in increased locomotor activity and reduced sleep, concordant with the human phenotype. CONCLUSIONS: This study reveals that a large set of intellectual disability-related genes contribute to ADHD risk through effects of common alleles. Utilizing this continuity, the authors identified TRAPPC9, MEF2C, and ST3GAL3 as novel ADHD candidate genes. Characterization in Drosophila suggests that TRAPPC9 and MEF2C contribute to ADHD-related behavior through distinct neural substrates.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Intercellular Signaling Peptides and Proteins/genetics , Locomotion/genetics , Myogenic Regulatory Factors/genetics , Sialyltransferases/genetics , Adult , Aged , Animals , Circadian Rhythm , Dopaminergic Neurons/metabolism , Female , Gene Knockdown Techniques , Genome-Wide Association Study , Humans , Intellectual Disability/genetics , MEF2 Transcription Factors/genetics , Male , Middle Aged , Neurons/metabolism , Sleep/genetics
18.
Neuropsychopharmacology ; 45(7): 1188-1195, 2020 06.
Article in English | MEDLINE | ID: mdl-31896117

ABSTRACT

Attention-Deficit/Hyperactivity Disorder (ADHD) and obesity are frequently comorbid, genetically correlated, and share brain substrates. The biological mechanisms driving this association are unclear, but candidate systems, like dopaminergic neurotransmission and circadian rhythm, have been suggested. Our aim was to identify the biological mechanisms underpinning the genetic link between ADHD and obesity measures and investigate associations of overlapping genes with brain volumes. We tested the association of dopaminergic and circadian rhythm gene sets with ADHD, body mass index (BMI), and obesity (using GWAS data of N = 53,293, N = 681,275, and N = 98,697, respectively). We then conducted genome-wide ADHD-BMI and ADHD-obesity gene-based meta-analyses, followed by pathway enrichment analyses. Finally, we tested the association of ADHD-BMI overlapping genes with brain volumes (primary GWAS data N = 10,720-10,928; replication data N = 9428). The dopaminergic gene set was associated with both ADHD (P = 5.81 × 10-3) and BMI (P = 1.63 × 10-5); the circadian rhythm was associated with BMI (P = 1.28 × 10-3). The genome-wide approach also implicated the dopaminergic system, as the Dopamine-DARPP32 Feedback in cAMP Signaling pathway was enriched in both ADHD-BMI and ADHD-obesity results. The ADHD-BMI overlapping genes were associated with putamen volume (P = 7.7 × 10-3; replication data P = 3.9 × 10-2)-a brain region with volumetric reductions in ADHD and BMI and linked to inhibitory control. Our findings suggest that dopaminergic neurotransmission, partially through DARPP-32-dependent signaling and involving the putamen, is a key player underlying the genetic overlap between ADHD and obesity measures. Uncovering shared etiological factors underlying the frequently observed ADHD-obesity comorbidity may have important implications in terms of prevention and/or efficient treatment of these conditions.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Dopamine , Attention Deficit Disorder with Hyperactivity/genetics , Body Mass Index , Dopamine/physiology , Humans , Obesity/genetics , Signal Transduction/genetics
20.
Sci Rep ; 8(1): 1881, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382897

ABSTRACT

Methylphenidate (MPH) is the most frequently used pharmacological treatment in children with attention-deficit/hyperactivity disorder (ADHD). However, a considerable interindividual variability exists in clinical outcome. Thus, we performed a genome-wide association study of MPH efficacy in 173 ADHD paediatric patients. Although no variant reached genome-wide significance, the set of genes containing single-nucleotide polymorphisms (SNPs) nominally associated with MPH response (P < 0.05) was significantly enriched for candidates previously studied in ADHD or treatment outcome. We prioritised the nominally significant SNPs by functional annotation and expression quantitative trait loci (eQTL) analysis in human brain, and we identified 33 SNPs tagging cis-eQTL in 32 different loci (referred to as eSNPs and eGenes, respectively). Pathway enrichment analyses revealed an over-representation of genes involved in nervous system development and function among the eGenes. Categories related to neurological diseases, psychological disorders and behaviour were also significantly enriched. We subsequently meta-analysed the association with clinical outcome for the 33 eSNPs across the discovery sample and an independent cohort of 189 ADHD adult patients (target sample) and we detected 15 suggestive signals. Following this comprehensive strategy, our results provide a better understanding of the molecular mechanisms implicated in MPH treatment effects and suggest promising candidates that may encourage future studies.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/genetics , Central Nervous System Stimulants/therapeutic use , Methylphenidate/therapeutic use , Adolescent , Child , Child, Preschool , Female , Genome-Wide Association Study , Genomics/methods , Humans , Male , Polymorphism, Single Nucleotide/drug effects , Polymorphism, Single Nucleotide/genetics , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...