Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Commun ; 15(1): 1770, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38413609

ABSTRACT

Common genetic variation has been associated with multiple phenotypic features in Autism Spectrum Disorder (ASD). However, our knowledge of shared genetic factor structures contributing to this highly heterogeneous phenotypic spectrum is limited. Here, we developed and implemented a structural equation modelling framework to directly model genomic covariance across core and non-core ASD phenotypes, studying autistic individuals of European descent with a case-only design. We identified three independent genetic factors most strongly linked to language performance, behaviour and developmental motor delay, respectively, studying an autism community sample (N = 5331). The three-factorial structure was largely confirmed in independent ASD-simplex families (N = 1946), although we uncovered, in addition, simplex-specific genetic overlap between behaviour and language phenotypes. Multivariate models across cohorts revealed novel associations, including links between language and early mastering of self-feeding. Thus, the common genetic architecture in ASD is multi-dimensional with overarching genetic factors contributing, in combination with ascertainment-specific patterns, to phenotypic heterogeneity.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Humans , Autistic Disorder/genetics , Autism Spectrum Disorder/genetics , Phenotype , Language , Models, Structural
2.
Proc Natl Acad Sci U S A ; 119(35): e2202764119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35998220

ABSTRACT

The use of spoken and written language is a fundamental human capacity. Individual differences in reading- and language-related skills are influenced by genetic variation, with twin-based heritability estimates of 30 to 80% depending on the trait. The genetic architecture is complex, heterogeneous, and multifactorial, but investigations of contributions of single-nucleotide polymorphisms (SNPs) were thus far underpowered. We present a multicohort genome-wide association study (GWAS) of five traits assessed individually using psychometric measures (word reading, nonword reading, spelling, phoneme awareness, and nonword repetition) in samples of 13,633 to 33,959 participants aged 5 to 26 y. We identified genome-wide significant association with word reading (rs11208009, P = 1.098 × 10-8) at a locus that has not been associated with intelligence or educational attainment. All five reading-/language-related traits showed robust SNP heritability, accounting for 13 to 26% of trait variability. Genomic structural equation modeling revealed a shared genetic factor explaining most of the variation in word/nonword reading, spelling, and phoneme awareness, which only partially overlapped with genetic variation contributing to nonword repetition, intelligence, and educational attainment. A multivariate GWAS of word/nonword reading, spelling, and phoneme awareness maximized power for follow-up investigation. Genetic correlation analysis with neuroimaging traits identified an association with the surface area of the banks of the left superior temporal sulcus, a brain region linked to the processing of spoken and written language. Heritability was enriched for genomic elements regulating gene expression in the fetal brain and in chromosomal regions that are depleted of Neanderthal variants. Together, these results provide avenues for deciphering the biological underpinnings of uniquely human traits.


Subject(s)
Genome-Wide Association Study , Individuality , Reading , Speech , Adolescent , Adult , Child , Child, Preschool , Genetic Loci , Humans , Language , Polymorphism, Single Nucleotide , Young Adult
3.
Mol Psychiatry ; 27(3): 1588-1598, 2022 03.
Article in English | MEDLINE | ID: mdl-35228676

ABSTRACT

Many mental health conditions present a spectrum of social difficulties that overlaps with social behaviour in the general population including shared but little characterised genetic links. Here, we systematically investigate heterogeneity in shared genetic liabilities with attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorders (ASD), bipolar disorder (BP), major depression (MD) and schizophrenia across a spectrum of different social symptoms. Longitudinally assessed low-prosociality and peer-problem scores in two UK population-based cohorts (4-17 years; parent- and teacher-reports; Avon Longitudinal Study of Parents and Children(ALSPAC): N ≤ 6,174; Twins Early Development Study(TEDS): N ≤ 7,112) were regressed on polygenic risk scores for disorder, as informed by genome-wide summary statistics from large consortia, using negative binomial regression models. Across ALSPAC and TEDS, we replicated univariate polygenic associations between social behaviour and risk for ADHD, MD and schizophrenia. Modelling variation in univariate genetic effects jointly using random-effect meta-regression revealed evidence for polygenic links between social behaviour and ADHD, ASD, MD, and schizophrenia risk, but not BP. Differences in age, reporter and social trait captured 45-88% in univariate effect variation. Cross-disorder adjusted analyses demonstrated that age-related heterogeneity in univariate effects is shared across mental health conditions, while reporter- and social trait-specific heterogeneity captures disorder-specific profiles. In particular, ADHD, MD, and ASD polygenic risk were more strongly linked to peer problems than low prosociality, while schizophrenia was associated with low prosociality only. The identified association profiles suggest differences in the social genetic architecture across mental disorders when investigating polygenic overlap with population-based social symptoms spanning 13 years of child and adolescent development.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Autism Spectrum Disorder , Adolescent , Attention Deficit Disorder with Hyperactivity/epidemiology , Autism Spectrum Disorder/epidemiology , Child , Genome-Wide Association Study , Humans , Longitudinal Studies , Multifactorial Inheritance/genetics , Social Behavior
4.
Mol Psychiatry ; 25(11): 3053-3065, 2020 11.
Article in English | MEDLINE | ID: mdl-30279459

ABSTRACT

The hippocampus is a heterogeneous structure, comprising histologically distinguishable subfields. These subfields are differentially involved in memory consolidation, spatial navigation and pattern separation, complex functions often impaired in individuals with brain disorders characterized by reduced hippocampal volume, including Alzheimer's disease (AD) and schizophrenia. Given the structural and functional heterogeneity of the hippocampal formation, we sought to characterize the subfields' genetic architecture. T1-weighted brain scans (n = 21,297, 16 cohorts) were processed with the hippocampal subfields algorithm in FreeSurfer v6.0. We ran a genome-wide association analysis on each subfield, co-varying for whole hippocampal volume. We further calculated the single-nucleotide polymorphism (SNP)-based heritability of 12 subfields, as well as their genetic correlation with each other, with other structural brain features and with AD and schizophrenia. All outcome measures were corrected for age, sex and intracranial volume. We found 15 unique genome-wide significant loci across six subfields, of which eight had not been previously linked to the hippocampus. Top SNPs were mapped to genes associated with neuronal differentiation, locomotor behaviour, schizophrenia and AD. The volumes of all the subfields were estimated to be heritable (h2 from 0.14 to 0.27, all p < 1 × 10-16) and clustered together based on their genetic correlations compared with other structural brain features. There was also evidence of genetic overlap of subicular subfield volumes with schizophrenia. We conclude that hippocampal subfields have partly distinct genetic determinants associated with specific biological processes and traits. Taking into account this specificity may increase our understanding of hippocampal neurobiology and associated pathologies.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Hippocampus/anatomy & histology , Hippocampus/pathology , Neuroimaging , Polymorphism, Single Nucleotide/genetics , Schizophrenia/genetics , Schizophrenia/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Child , Child, Preschool , Female , Genome-Wide Association Study , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Humans , Male , Middle Aged , Schizophrenia/diagnostic imaging , Young Adult
5.
Eur Neuropsychopharmacol ; 30: 44-55, 2020 01.
Article in English | MEDLINE | ID: mdl-29174947

ABSTRACT

The RBFOX1 gene (or A2BP1) encodes a splicing factor important for neuronal development that has been related to autism spectrum disorder and other neurodevelopmental phenotypes. Evidence from complementary sources suggests that this gene contributes to aggressive behavior. Suggestive associations with RBFOX1 have been identified in genome-wide association studies (GWAS) of anger, conduct disorder, and aggressive behavior. Nominal association signals in RBFOX1 were also found in an epigenome-wide association study (EWAS) of aggressive behavior. Also, variants in this gene affect temporal lobe volume, a brain area that is altered in several aggression-related phenotypes. In animals, this gene has been shown to modulate aggressive behavior in Drosophila. RBFOX1 has also been associated with canine aggression and is upregulated in mice that show increased aggression after frustration of an expected reward. Associated common genetic variants as well as rare duplications and deletions affecting RBFOX1 have been identified in several psychiatric and neurodevelopmental disorders that are often comorbid with aggressive behaviors. In this paper, we comprehensively review the cumulative evidence linking RBFOX1 to aggression behavior and provide new results implicating RBFOX1 in this phenotype. Most of these studies (genetic and epigenetic analyses in humans, neuroimaging genetics, gene expression and animal models) are hypothesis-free, which strengthens the validity of the findings, although all the evidence is nominal and should therefore be taken with caution. Further studies are required to clarify in detail the role of this gene in this complex phenotype.


Subject(s)
Aggression/physiology , Aggression/psychology , Genetic Association Studies/methods , RNA Splicing Factors/genetics , Animals , Epigenesis, Genetic/physiology , Genetic Variation/physiology , Genome-Wide Association Study/methods , Humans , RNA Splicing Factors/biosynthesis
6.
Eur Neuropsychopharmacol ; 30: 75-86, 2020 01.
Article in English | MEDLINE | ID: mdl-29191428

ABSTRACT

Investigating phenotypic heterogeneity in aggression and understanding the molecular biological basis of aggression subtypes may lead to new prevention and treatment options. In the current study, we evaluated the taxonomy of aggression and examined specific genetic mechanisms underlying aggression subtypes in healthy males and females. Confirmatory Factor Analysis (CFA) was used to replicate a recently reported three-factor model of the Reactive Proactive Questionnaire (RPQ) in healthy adults (n = 661; median age 24.0 years; 41% male). Gene-set association analysis, aggregating common genetic variants within (a combination of) three molecular pathways previously implicated in aggression, i.e. serotonergic, dopaminergic, and neuroendocrine signaling, was conducted with MAGMA software in males and females separately (total n = 395) for aggression subtypes. We replicate the three-factor CFA model of the RPQ, and found males to score significantly higher on one of these factors compared to females: proactive aggression. The genetic association analysis showed a female-specific association of genetic variation in the combined gene-set with a different factor of the RPQ; reactive aggression due to internal frustration. Both the neuroendocrine and serotonergic gene-sets contributed significantly to this association. Our genetic findings are subtype- and sex-specific, stressing the value of efforts to reduce heterogeneity in research of aggression etiology. Importantly, subtype- and sex-differences in the underlying pathophysiology of aggression suggest that optimal treatment options will have to be tailored to the individual patient. Male and female needs of intervention might differ, stressing the need for sex-specific further research of aggression. Our work highlights opportunities for sample size maximization offered by population-based studies of aggression.


Subject(s)
Aggression/physiology , Biogenic Monoamines/physiology , Frustration , Genetic Association Studies/methods , Neurosecretory Systems/physiology , Sex Characteristics , Adult , Aggression/psychology , Factor Analysis, Statistical , Female , Humans , Male , Surveys and Questionnaires , Young Adult
7.
Hum Brain Mapp ; 40(18): 5202-5212, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31441562

ABSTRACT

Previous studies have linked the low expression variant of a variable number of tandem repeat polymorphism in the monoamine oxidase A gene (MAOA-L) to the risk for impulsivity and aggression, brain developmental abnormalities, altered cortico-limbic circuit function, and an exaggerated neural serotonergic tone. However, the neurobiological effects of this variant on human brain network architecture are incompletely understood. We studied healthy individuals and used multimodal neuroimaging (sample size range: 219-284 across modalities) and network-based statistics (NBS) to probe the specificity of MAOA-L-related connectomic alterations to cortical-limbic circuits and the emotion processing domain. We assessed the spatial distribution of affected links across several neuroimaging tasks and data modalities to identify potential alterations in network architecture. Our results revealed a distributed network of node links with a significantly increased connectivity in MAOA-L carriers compared to the carriers of the high expression (H) variant. The hyperconnectivity phenotype primarily consisted of between-lobe ("anisocoupled") network links and showed a pronounced involvement of frontal-temporal connections. Hyperconnectivity was observed across functional magnetic resonance imaging (fMRI) of implicit emotion processing (pFWE = .037), resting-state fMRI (pFWE = .022), and diffusion tensor imaging (pFWE = .044) data, while no effects were seen in fMRI data of another cognitive domain, that is, spatial working memory (pFWE = .540). These observations are in line with prior research on the MAOA-L variant and complement these existing data by novel insights into the specificity and spatial distribution of the neurogenetic effects. Our work highlights the value of multimodal network connectomic approaches for imaging genetics.


Subject(s)
Brain/diagnostic imaging , Genotype , Magnetic Resonance Imaging/methods , Minisatellite Repeats/genetics , Monoamine Oxidase/genetics , Nerve Net/diagnostic imaging , Adult , Brain/physiology , Female , Frontal Lobe/diagnostic imaging , Frontal Lobe/physiology , Humans , Male , Nerve Net/physiology , Temporal Lobe/diagnostic imaging , Temporal Lobe/physiology , Young Adult
8.
Dev Sci ; 22(3): e12763, 2019 05.
Article in English | MEDLINE | ID: mdl-30318656

ABSTRACT

Given the long-lasting detrimental effects of internalizing symptoms, there is great need for detecting early risk markers. One promising marker is freezing behavior. Whereas initial freezing reactions are essential for coping with threat, prolonged freezing has been associated with internalizing psychopathology. However, it remains unknown whether early life alterations in freezing reactions predict changes in internalizing symptoms during adolescent development. In a longitudinal study (N = 116), we tested prospectively whether observed freezing in infancy predicted the development of internalizing symptoms from childhood through late adolescence (until age 17). Both longer and absent infant freezing behavior during a standard challenge (robot-confrontation task) were associated with internalizing symptoms in adolescence. Specifically, absent infant freezing predicted a relative increase in internalizing symptoms consistently across development from relatively low symptom levels in childhood to relatively high levels in late adolescence. Longer infant freezing also predicted a relative increase in internalizing symptoms, but only up until early adolescence. This latter effect was moderated by peer stress and was followed by a later decrease in internalizing symptoms. The findings suggest that early deviations in defensive freezing responses signal risk for internalizing symptoms and may constitute important markers in future stress vulnerability and resilience studies.


Subject(s)
Adaptation, Psychological/physiology , Adolescent Development/physiology , Fear/psychology , Stress, Psychological/psychology , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Longitudinal Studies , Male , Peer Group , Polymorphism, Genetic/genetics , Prospective Studies , Serotonin Plasma Membrane Transport Proteins/genetics
9.
Front Behav Neurosci ; 12: 61, 2018.
Article in English | MEDLINE | ID: mdl-29666571

ABSTRACT

Reactive and proactive subtypes of aggression have been recognized to help parse etiological heterogeneity of this complex phenotype. With a heritability of about 50%, genetic factors play a role in the development of aggressive behavior. Imaging studies implicate brain structures related to social behavior in aggression etiology, most notably the amygdala and striatum. This study aimed to gain more insight into the pathways from genetic risk factors for aggression to aggression phenotypes. To this end, we conducted genome-wide gene-based cross-trait meta-analyses of aggression with the volumes of amygdala, nucleus accumbens and caudate nucleus to identify genes influencing both aggression and aggression-related brain volumes. We used data of large-scale genome-wide association studies (GWAS) of: (a) aggressive behavior in children and adolescents (EAGLE, N = 18,988); and (b) Magnetic Resonance Imaging (MRI)-based volume measures of aggression-relevant subcortical brain regions (ENIGMA2, N = 13,171). Second, the identified genes were further investigated in a sample of healthy adults (mean age (SD) = 25.28 (4.62) years; 43% male) who had genome-wide genotyping data and questionnaire data on aggression subtypes available (Brain Imaging Genetics, BIG, N = 501) to study their effect on reactive and proactive subtypes of aggression. Our meta-analysis identified two genes, MECOM and AVPR1A, significantly associated with both aggression risk and nucleus accumbens (MECOM) and amygdala (AVPR1A) brain volume. Subsequent in-depth analysis of these genes in healthy adults (BIG), including sex as an interaction term in the model, revealed no significant subtype-specific gene-wide associations. Using cross-trait meta-analysis of brain measures and psychiatric phenotypes, this study generated new hypotheses about specific links between genes, the brain and behavior. Results indicate that MECOM and AVPR1A may exert an effect on aggression through mechanisms involving nucleus accumbens and amygdala volumes, respectively.

10.
Neurosci Biobehav Rev ; 80: 115-155, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28159610

ABSTRACT

Attention-deficit/hyperactivity disorder (ADHD) is a common and often persistent neurodevelopmental disorder. Beyond gene-finding, neurobiological parameters, such as brain structure, connectivity, and function, have been used to link genetic variation to ADHD symptomatology. We performed a systematic review of brain imaging genetics studies involving 62 ADHD candidate genes in childhood and adult ADHD cohorts. Fifty-one eligible research articles described studies of 13 ADHD candidate genes. Almost exclusively, single genetic variants were studied, mostly focussing on dopamine-related genes. While promising results have been reported, imaging genetics studies are thus far hampered by methodological differences in study design and analysis methodology, as well as limited sample sizes. Beyond reviewing imaging genetics studies, we also discuss the need for complementary approaches at multiple levels of biological complexity and emphasize the importance of combining and integrating findings across levels for a better understanding of biological pathways from gene to disease. These may include multi-modal imaging genetics studies, bioinformatic analyses, and functional analyses of cell and animal models.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Attention Deficit Disorder with Hyperactivity/physiopathology , Neural Pathways/physiopathology , Neuroimaging , Attention Deficit Disorder with Hyperactivity/pathology , Genetic Predisposition to Disease/genetics , Humans , Neural Pathways/pathology
11.
Am J Med Genet B Neuropsychiatr Genet ; 174(5): 485-537, 2017 07.
Article in English | MEDLINE | ID: mdl-29984470

ABSTRACT

Neurodevelopmental disorders are defined by highly heritable problems during development and brain growth. Attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorders (ASDs), and intellectual disability (ID) are frequent neurodevelopmental disorders, with common comorbidity among them. Imaging genetics studies on the role of disease-linked genetic variants on brain structure and function have been performed to unravel the etiology of these disorders. Here, we reviewed imaging genetics literature on these disorders attempting to understand the mechanisms of individual disorders and their clinical overlap. For ADHD and ASD, we selected replicated candidate genes implicated through common genetic variants. For ID, which is mainly caused by rare variants, we included genes for relatively frequent forms of ID occurring comorbid with ADHD or ASD. We reviewed case-control studies and studies of risk variants in healthy individuals. Imaging genetics studies for ADHD were retrieved for SLC6A3/DAT1, DRD2, DRD4, NOS1, and SLC6A4/5HTT. For ASD, studies on CNTNAP2, MET, OXTR, and SLC6A4/5HTT were found. For ID, we reviewed the genes FMR1, TSC1 and TSC2, NF1, and MECP2. Alterations in brain volume, activity, and connectivity were observed. Several findings were consistent across studies, implicating, for example, SLC6A4/5HTT in brain activation and functional connectivity related to emotion regulation. However, many studies had small sample sizes, and hypothesis-based, brain region-specific studies were common. Results from available studies confirm that imaging genetics can provide insight into the link between genes, disease-related behavior, and the brain. However, the field is still in its early stages, and conclusions about shared mechanisms cannot yet be drawn.


Subject(s)
Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/genetics , Psychopathology/methods , Attention Deficit Disorder with Hyperactivity/etiology , Attention Deficit Disorder with Hyperactivity/genetics , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/genetics , Brain/pathology , Case-Control Studies , Comorbidity , Genetic Predisposition to Disease , Humans , Intellectual Disability/etiology , Intellectual Disability/genetics , Risk Factors
12.
Am J Med Genet B Neuropsychiatr Genet ; 171(5): 733-47, 2016 07.
Article in English | MEDLINE | ID: mdl-27021288

ABSTRACT

Aggressiveness is a behavioral trait that has the potential to be harmful to individuals and society. With an estimated heritability of about 40%, genetics is important in its development. We performed an exploratory genome-wide association (GWA) analysis of childhood aggressiveness in attention deficit hyperactivity disorder (ADHD) to gain insight into the underlying biological processes associated with this trait. Our primary sample consisted of 1,060 adult ADHD patients (aADHD). To further explore the genetic architecture of childhood aggressiveness, we performed enrichment analyses of suggestive genome-wide associations observed in aADHD among GWA signals of dimensions of oppositionality (defiant/vindictive and irritable dimensions) in childhood ADHD (cADHD). No single polymorphism reached genome-wide significance (P < 5.00E-08). The strongest signal in aADHD was observed at rs10826548, within a long noncoding RNA gene (beta = -1.66, standard error (SE) = 0.34, P = 1.07E-06), closely followed by rs35974940 in the neurotrimin gene (beta = 3.23, SE = 0.67, P = 1.26E-06). The top GWA SNPs observed in aADHD showed significant enrichment of signals from both the defiant/vindictive dimension (Fisher's P-value = 2.28E-06) and the irritable dimension in cADHD (Fisher's P-value = 0.0061). In sum, our results identify a number of biologically interesting markers possibly underlying childhood aggressiveness and provide targets for further genetic exploration of aggressiveness across psychiatric disorders. © 2016 The Authors. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics Published by Wiley Periodicals, Inc.


Subject(s)
Aggression/physiology , Attention Deficit Disorder with Hyperactivity/genetics , Adolescent , Adult , Aggression/psychology , Attention Deficit Disorder with Hyperactivity/complications , Attention Deficit Disorder with Hyperactivity/psychology , Child , Female , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Humans , Male , Polymorphism, Single Nucleotide/genetics
13.
Am J Med Genet B Neuropsychiatr Genet ; 171(5): 573-88, 2016 07.
Article in English | MEDLINE | ID: mdl-26184070

ABSTRACT

Oppositional defiant disorder (ODD) is a frequent psychiatric disorder seen in children and adolescents with attention-deficit-hyperactivity disorder (ADHD). ODD is also a common antecedent to both affective disorders and aggressive behaviors. Although the heritability of ODD has been estimated to be around 0.60, there has been little research into the molecular genetics of ODD. The present study examined the association of irritable and defiant/vindictive dimensions and categorical subtypes of ODD (based on latent class analyses) with previously described specific polymorphisms (DRD4 exon3 VNTR, 5-HTTLPR, and seven OXTR SNPs) as well as with dopamine, serotonin, and oxytocin genes and pathways in a clinical sample of children and adolescents with ADHD. In addition, we performed a multivariate genome-wide association study (GWAS) of the aforementioned ODD dimensions and subtypes. Apart from adjusting the analyses for age and sex, we controlled for "parental ability to cope with disruptive behavior." None of the hypothesis-driven analyses revealed a significant association with ODD dimensions and subtypes. Inadequate parenting behavior was significantly associated with all ODD dimensions and subtypes, most strongly with defiant/vindictive behaviors. In addition, the GWAS did not result in genome-wide significant findings but bioinformatics and literature analyses revealed that the proteins encoded by 28 of the 53 top-ranked genes functionally interact in a molecular landscape centered around Beta-catenin signaling and involved in the regulation of neurite outgrowth. Our findings provide new insights into the molecular basis of ODD and inform future genetic studies of oppositional behavior. © 2015 The Authors. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics Published by Wiley Periodicals, Inc.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Attention Deficit and Disruptive Behavior Disorders/genetics , Attention Deficit and Disruptive Behavior Disorders/psychology , Adolescent , Aggression/psychology , Attention Deficit Disorder with Hyperactivity/psychology , Child , Female , Genetic Association Studies/methods , Genome-Wide Association Study , Humans , Male , Parenting/psychology , Parents , Polymorphism, Single Nucleotide/genetics
14.
Addict Biol ; 21(4): 915-23, 2016 07.
Article in English | MEDLINE | ID: mdl-25752199

ABSTRACT

Individuals with attention deficit/hyperactivity disorder (ADHD) are at increased risk of developing substance use disorders (SUDs) and nicotine dependence. The co-occurrence of ADHD and SUDs/nicotine dependence may in part be mediated by shared genetic liability. Several neurobiological pathways have been implicated in both ADHD and SUDs, including dopamine and serotonin pathways. We hypothesized that variations in dopamine and serotonin neurotransmission genes were involved in the genetic liability to develop SUDs/nicotine dependence in ADHD. The current study included participants with ADHD (n = 280) who were originally part of the Dutch International Multicenter ADHD Genetics study. Participants were aged 5-15 years and attending outpatient clinics at enrollment in the study. Diagnoses of ADHD, SUDs, nicotine dependence, age of first nicotine and substance use, and alcohol use severity were based on semi-structured interviews and questionnaires. Genetic risk scores were created for both serotonergic and dopaminergic risk genes previously shown to be associated with ADHD and SUDs and/or nicotine dependence. The serotonin genetic risk score significantly predicted alcohol use severity. No significant serotonin × dopamine risk score or effect of stimulant medication was found. The current study adds to the literature by providing insight into genetic underpinnings of the co-morbidity of ADHD and SUDs. While the focus of the literature so far has been mostly on dopamine, our study suggests that serotonin may also play a role in the relationship between these disorders.


Subject(s)
Attention Deficit Disorder with Hyperactivity/epidemiology , Dopamine/genetics , Genetic Predisposition to Disease/epidemiology , Serotonin/genetics , Substance-Related Disorders/epidemiology , Substance-Related Disorders/genetics , Adolescent , Adult , Attention Deficit Disorder with Hyperactivity/genetics , Comorbidity , Female , Follow-Up Studies , Genetic Predisposition to Disease/genetics , Humans , Male , Netherlands/epidemiology , Risk Factors , Surveys and Questionnaires , Tobacco Use Disorder/epidemiology , Tobacco Use Disorder/genetics , Young Adult
15.
World J Biol Psychiatry ; 16(8): 625-34, 2015.
Article in English | MEDLINE | ID: mdl-26428549

ABSTRACT

OBJECTIVES: Deficits in response inhibition have been associated with attention-deficit/hyperactivity disorder (ADHD). Given the role of serotonin in ADHD and impulsivity, we postulated that genetic variants within the serotonin pathway might influence response inhibition. METHODS: We measured neural activation during stop-signal task performance in adolescents with ADHD (N = 185), their unaffected siblings (N = 111), and healthy controls (N = 124), and investigated the relationship of two serotonin gene polymorphisms (the rs6296 SNP of the HTR1B gene and HTTLPR variants of the 5-HTT gene) with the neural correlates of response inhibition. RESULTS: The whole-brain analyses demonstrated large scale neural activation differences in the inferior and medial frontal and temporal/parietal regions of the response inhibition network between the different variants of both the HTR1B and 5HTT genes. Activation in these regions was significantly associated with stop-task performance, but not with ADHD diagnosis or severity. No associations were found between HTR1B and 5HTT variants and ADHD or ADHD-related neural activation. CONCLUSIONS: These results provide novel evidence that serotonin may play an important role in the neurobiology of response inhibition. Although response inhibition is strongly linked to ADHD, serotonin linked genetic variants associated with response inhibition and its neural correlates do not explain variance of the ADHD phenotype.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Parietal Lobe/pathology , Receptor, Serotonin, 5-HT1B/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Synaptic Transmission/genetics , Adolescent , Adult , Attention , Attention Deficit Disorder with Hyperactivity/diagnosis , Brain Mapping , Case-Control Studies , Female , Humans , Impulsive Behavior , Magnetic Resonance Imaging , Male , Netherlands , Neuropsychological Tests , Task Performance and Analysis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...