51
|
Mehta T, Mannem N, Yarasi NK, Bollu PC. Biomarkers for ADHD: the Present and Future Directions. CURRENT DEVELOPMENTAL DISORDERS REPORTS 2020. [DOI: 10.1007/s40474-020-00196-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
52
|
Esteller-Cucala P, Maceda I, Børglum AD, Demontis D, Faraone SV, Cormand B, Lao O. Genomic analysis of the natural history of attention-deficit/hyperactivity disorder using Neanderthal and ancient Homo sapiens samples. Sci Rep 2020; 10:8622. [PMID: 32451437 PMCID: PMC7248073 DOI: 10.1038/s41598-020-65322-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/24/2020] [Indexed: 11/18/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is an impairing neurodevelopmental condition highly prevalent in current populations. Several hypotheses have been proposed to explain this paradox, mainly in the context of the Paleolithic versus Neolithic cultural shift but especially within the framework of the mismatch theory. This theory elaborates on how a particular trait once favoured in an ancient environment might become maladaptive upon environmental changes. However, given the lack of genomic data available for ADHD, these theories have not been empirically tested. We took advantage of the largest GWAS meta-analysis available for this disorder consisting of over 20,000 individuals diagnosed with ADHD and 35,000 controls, to assess the evolution of ADHD-associated alleles in European populations using archaic, ancient and modern human samples. We also included Approximate Bayesian computation coupled with deep learning analyses and singleton density scores to detect human adaptation. Our analyses indicate that ADHD-associated alleles are enriched in loss of function intolerant genes, supporting the role of selective pressures in this early-onset phenotype. Furthermore, we observed that the frequency of variants associated with ADHD has steadily decreased since Paleolithic times, particularly in Paleolithic European populations compared to samples from the Neolithic Fertile Crescent. We demonstrate this trend cannot be explained by African admixture nor Neanderthal introgression, since introgressed Neanderthal alleles are enriched in ADHD risk variants. All analyses performed support the presence of long-standing selective pressures acting against ADHD-associated alleles until recent times. Overall, our results are compatible with the mismatch theory for ADHD but suggest a much older time frame for the evolution of ADHD-associated alleles compared to previous hypotheses.
Collapse
Affiliation(s)
- Paula Esteller-Cucala
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institut de Biologia Evolutiva (UPF-CSIC), Barcelona, Spain
| | - Iago Maceda
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Anders D Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, and Aarhus Genome Centre, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Ditte Demontis
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, and Aarhus Genome Centre, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain.
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Spain.
| | - Oscar Lao
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
53
|
Song Y, Lu M, Yuan H, Chen T, Han X. Mast cell-mediated neuroinflammation may have a role in attention deficit hyperactivity disorder (Review). Exp Ther Med 2020; 20:714-726. [PMID: 32742317 PMCID: PMC7388140 DOI: 10.3892/etm.2020.8789] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a common neurodevelopmental and behavioral disorder with a serious negative impact on the quality of life from childhood until adulthood, which may cause academic failure, family disharmony and even social unrest. The pathogenesis of ADHD has remained to be fully elucidated, leading to difficulties in the treatment of this disease. Genetic and environmental factors contribute to the risk of ADHD development. Certain studies indicated that ADHD has high comorbidity with allergic and autoimmune diseases, with various patients with ADHD having a high inflammatory status. Increasing evidence indicated that mast cells (MCs) are involved in the pathogenesis of brain inflammation and neuropsychiatric disorders. MCs may cause or aggravate neuroinflammation via the selective release of inflammatory factors, interaction with glial cells and neurons, activation of the hypothalamic-pituitary adrenal axis or disruption of the blood-brain barrier integrity. In the present review, the notion that MC activation may be involved in the occurrence and development of ADHD through a number of ways is discussed based on previously published studies. The association between MCs and ADHD appears to lack sufficient evidence at present and this hypothesis is considered to be worthy of further study, providing a novel perspective for the treatment of ADHD.
Collapse
Affiliation(s)
- Yuchen Song
- Institute of Pediatrics of Traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Manqi Lu
- Institute of Pediatrics of Traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Haixia Yuan
- Institute of Pediatrics of Traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Tianyi Chen
- Institute of Pediatrics of Traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xinmin Han
- Institute of Pediatrics of Traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
54
|
Baccarin M, Picinelli C, Tomaiuolo P, Castronovo P, Costa A, Verdecchia M, Cannizzaro C, Barbieri G, Sacco R, Persico AM, Lintas C. Appropriateness of array-CGH in the ADHD clinics: A comparative study. GENES BRAIN AND BEHAVIOR 2020; 19:e12651. [PMID: 32141190 DOI: 10.1111/gbb.12651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 01/24/2023]
Abstract
Attention deficit hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorder with a worldwide prevalence of about 5%. The disorder is characterized by inattentive, hyperactive and impulsive behavior and is often comorbid with other neuropsychiatric conditions. Array comparative genomic hybridization (array-CGH) testing has been proved to be useful to detect chromosomal aberrations in several neuropsychiatric conditions including autism spectrum disorders (ASD) and intellectual disability (ID). The usefulness of array-CGH in the ADHD clinics is still debated and no conclusive evidence has been reached to date. We performed array-CGH in 98 children and adolescents divided in two similarly sized groups according to the clinical diagnosis: (a) one group diagnosed with ADHD as primary diagnosis; (b) the other group in which ADHD was co-morbid with ASD and/or ID. We detected pathogenetic and likely pathogenetic copy number variants (CNVs) in 12% subjects in which ADHD was co-morbid with autism and/or intellectual disability and in 8.5% subjects diagnosed with ADHD as primary diagnosis. Detection of CNVs of unknown clinical significance was similar in the two groups being 27% and 32%, respectively. Benign and likely benign CNVs accounted for 61% and 59.5% in the first and second group, respectively. Differences in the diagnostic yield were not statistically significant between the two groups (P > .05). Our data strongly suggest that array-CGH (a) is a valuable diagnostic tool to detect clinically significant CNVs in individuals with ADHD even in the absence of comorbidity with ASD and/or ID and (b) should be implemented routinely in the ADHD clinics.
Collapse
Affiliation(s)
- Marco Baccarin
- Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| | - Chiara Picinelli
- Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| | | | - Paola Castronovo
- Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| | - Anna Costa
- Service for Neurodevelopmental Disorders, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Magda Verdecchia
- Service for Neurodevelopmental Disorders, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Chiara Cannizzaro
- Service for Neurodevelopmental Disorders, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Giusi Barbieri
- Service for Neurodevelopmental Disorders, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Roberto Sacco
- Service for Neurodevelopmental Disorders, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Antonio M Persico
- Interdepartmental Program "Autism 0-90", "Gaetano Martino" University Hospital, University of Messina, Messina, Italy
| | - Carla Lintas
- Service for Neurodevelopmental Disorders, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| |
Collapse
|
55
|
Bahn GH, Lee YS, Yoo HK, Kim EJ, Park S, Han DH, Hong M, Kim B, Lee SI, Bhang SY, Lee SY, Hong JP, Joung YS. Development of the Korean Practice Parameter for Adult Attention-Deficit/Hyperactivity Disorder. Soa Chongsonyon Chongsin Uihak 2020; 31:5-25. [PMID: 32612409 PMCID: PMC7324844 DOI: 10.5765/jkacap.190030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES Adult attention-deficit/hyperactivity disorder (ADHD) is an important mental health problem that needs resolution, especially considering the high rates of ADHD continuation from childhood to adolescence/adulthood and the high prevalence of ADHD in adults. Adults with ADHD have lifelong negative impacts and require close monitoring with long-term follow-up. Hence, the establishment of a Korean practice parameter for adult ADHD is necessary to minimize discontinuation of treatment and enable information sharing among Korean mental health professionals. METHODS The Korean practice parameter was developed using an evidence-based approach consisting of expert consensus survey coupled with literature review. RESULTS According to the expert consensus survey, the most commonly used diagnostic methods were clinical psychiatric interview (20.66%) and self-report scales (19.25%) followed by attention (14.71%) and psychological tests (14.24%). Key evaluation instruments currently available in Korea are the World Health Organization Adult ADHD Self-Report Rating Scale, Korean Adult ADHD Rating Scale, Diagnostic Interview for ADHD in Adults, Barkley Deficits in Executive Functioning Scale for adults, Comprehensive Attention Test, Conners' Continuous Performance Test, and the subtests of Wechsler Adult Intelligence Scale, Digit Span and Letter-Number Sequencing. Although pharmacotherapy is recommended as the first-line of treatment for adult ADHD, we recommend that it be followed by a multimodal and multidisciplinary approach including psychoeducation, pharmacotherapy, cognitive behavior therapy and coaching. CONCLUSION The Korean practice parameter introduces not only general information for the diagnosis and treatment of adult ADHD on a global scale, but also the process of diagnosis and treatment options tailored to the Korean population.
Collapse
Affiliation(s)
- Geon Ho Bahn
- Department of Psychiatry, Kyung Hee University School of Medicine, Seoul, Korea
| | - Young Sik Lee
- Department of Psychiatry, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| | | | - Eui-Jung Kim
- Department of Psychiatry, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Subin Park
- Department of Research Planning, Mental Health Research Institute, National Center for Mental Health, Seoul, Korea
| | - Doug Hyun Han
- Department of Psychiatry, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| | - Minha Hong
- Department of Psychiatry, Myongji Hospital, Hanyang University College of Medicine, Seoul, Korea
| | - Bongseog Kim
- Department of Psychiatry, Inje University College of Medicine, Seoul, Korea
| | - Soyoung Irene Lee
- Department of Psychiatry, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Buchun, Korea
| | - Soo Young Bhang
- Department of Psychiatry, Eulji University School of Medicine, Seoul, Korea
| | - Seung Yup Lee
- Department of Psychiatry, Kyung Hee University School of Medicine, Seoul, Korea
| | - Jin Pyo Hong
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yoo-Sook Joung
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
56
|
CK1δ over-expressing mice display ADHD-like behaviors, frontostriatal neuronal abnormalities and altered expressions of ADHD-candidate genes. Mol Psychiatry 2020; 25:3322-3336. [PMID: 31363163 PMCID: PMC7714693 DOI: 10.1038/s41380-018-0233-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 07/04/2018] [Accepted: 07/18/2018] [Indexed: 11/08/2022]
Abstract
The cognitive mechanisms underlying attention-deficit hyperactivity disorder (ADHD), a highly heritable disorder with an array of candidate genes and unclear genetic architecture, remain poorly understood. We previously demonstrated that mice overexpressing CK1δ (CK1δ OE) in the forebrain show hyperactivity and ADHD-like pharmacological responses to D-amphetamine. Here, we demonstrate that CK1δ OE mice exhibit impaired visual attention and a lack of D-amphetamine-induced place preference, indicating a disruption of the dopamine-dependent reward pathway. We also demonstrate the presence of abnormalities in the frontostriatal circuitry, differences in synaptic ultra-structures by electron microscopy, as well as electrophysiological perturbations of both glutamatergic and GABAergic transmission, as observed by altered frequency and amplitude of mEPSCs and mIPSCs. Furthermore, gene expression profiling by next-generation sequencing alone, or in combination with bacTRAP technology to study specifically Drd1a versus Drd2 medium spiny neurons, revealed that developmental CK1δ OE alters transcriptional homeostasis in the striatum, including specific alterations in Drd1a versus Drd2 neurons. These results led us to perform a fine molecular characterization of targeted gene networks and pathway analysis. Importantly, a large fraction of 92 genes identified by GWAS studies as associated with ADHD in humans are significantly altered in our mouse model. The multiple abnormalities described here might be responsible for synaptic alterations and lead to complex behavioral abnormalities. Collectively, CK1δ OE mice share characteristics typically associated with ADHD and should represent a valuable model to investigate the disease in vivo.
Collapse
|
57
|
Examining overlap and homogeneity in ASD, ADHD, and OCD: a data-driven, diagnosis-agnostic approach. Transl Psychiatry 2019; 9:318. [PMID: 31772171 PMCID: PMC6880188 DOI: 10.1038/s41398-019-0631-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 10/04/2019] [Accepted: 10/20/2019] [Indexed: 12/18/2022] Open
Abstract
The validity of diagnostic labels of autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), and obsessive compulsive disorder (OCD) is an open question given the mounting evidence that these categories may not correspond to conditions with distinct etiologies, biologies, or phenotypes. The objective of this study was to determine the agreement between existing diagnostic labels and groups discovered based on a data-driven, diagnosis-agnostic approach integrating cortical neuroanatomy and core-domain phenotype features. A machine learning pipeline, called bagged-multiview clustering, was designed to discover homogeneous subgroups by integrating cortical thickness data and measures of core-domain phenotypic features of ASD, ADHD, and OCD. This study was conducted using data from the Province of Ontario Neurodevelopmental Disorders (POND) Network, a multi-center study in Ontario, Canada. Participants (n = 226) included children between the ages of 6 and 18 with a diagnosis of ASD (n = 112, median [IQR] age = 11.7[4.8], 21% female), ADHD (n = 58, median [IQR] age = 10.2[3.3], 14% female), or OCD (n = 34, median [IQR] age = 12.1[4.2], 38% female), as well as typically developing controls (n = 22, median [IQR] age = 11.0[3.8], 55% female). The diagnosis-agnostic groups were significantly different than each other in phenotypic characteristics (SCQ: χ2(9) = 111.21, p < 0.0001; SWAN: χ2(9) = 142.44, p < 0.0001) as well as cortical thickness in 75 regions of the brain. The analyses revealed disagreement between existing diagnostic labels and the diagnosis-agnostic homogeneous groups (normalized mutual information < 0.20). Our results did not support the validity of existing diagnostic labels of ASD, ADHD, and OCD as distinct entities with respect to phenotype and cortical morphology.
Collapse
|
58
|
Stobernack T, de Vries SPW, Rodrigues Pereira R, Pelsser LM, Ter Braak CJF, Aarts E, van Baarlen P, Kleerebezem M, Frankena K, Hontelez S. Biomarker Research in ADHD: the Impact of Nutrition (BRAIN) - study protocol of an open-label trial to investigate the mechanisms underlying the effects of a few-foods diet on ADHD symptoms in children. BMJ Open 2019; 9:e029422. [PMID: 31694844 PMCID: PMC6858247 DOI: 10.1136/bmjopen-2019-029422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/02/2019] [Accepted: 09/12/2019] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Attention deficit hyperactivity disorder (ADHD) is the most common childhood behavioural disorder, causing significant impediment to a child's development. It is a complex disorder with numerous contributing (epi)genetic and environmental factors. Currently, treatment consists of behavioural and pharmacological therapy. However, ADHD medication is associated with several side effects, and concerns about long-term effects and efficacy exist. Therefore, there is considerable interest in the development of alternative treatment options. Double-blind research investigating the effects of a few-foods diet (FFD) has demonstrated a significant decrease in ADHD symptoms following an FFD. However, an FFD requires a considerable effort of both child and parents, limiting its applicability as a general ADHD treatment. To make FFD intervention less challenging or potentially obsolete, we need to understand how, and in which children, an FFD affects ADHD behaviour and, consequently, the child's well-being. We hypothesise that an FFD affects brain function, and that the nutritional impact on ADHD is effectuated by a complex interplay between the microbiota, gut and brain, that is, the microbiota-gut-brain axis. METHODS AND ANALYSIS The Biomarker Research in ADHD: the Impact of Nutrition (BRAIN) study is an open-label trial with researchers blinded to changes in ADHD symptoms during sample processing and initial data analyses. ETHICS AND DISSEMINATION The Medical Research and Ethics Committee of Wageningen University has approved this study (NL63851.081.17, application 17/24). Results will be disseminated through peer-reviewed journal publications, conference presentations, (social) media and the BRAIN study website. A summary of the findings will be provided to the participants. TRIAL REGISTRATION NUMBER NCT03440346. STUDY DATES Collection of primary outcome data started in March 2018 and will be ongoing until 100 children have participated in the study. Sample data analysis will start after all samples have been collected.
Collapse
Affiliation(s)
- Tim Stobernack
- Host-Microbe Interactomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Stefan P W de Vries
- Host-Microbe Interactomics, Wageningen University and Research, Wageningen, The Netherlands
| | | | | | - Cajo J F Ter Braak
- Biometris, Wageningen University and Research, Wageningen, The Netherlands
| | - Esther Aarts
- Donders Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, The Netherlands
| | - Peter van Baarlen
- Host-Microbe Interactomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Michiel Kleerebezem
- Host-Microbe Interactomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Klaas Frankena
- Adaptation Physiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Saartje Hontelez
- Host-Microbe Interactomics, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
59
|
Parent SNAP‐IV rating of attention‐deficit/hyperactivity disorder: accuracy in a clinical sample of ADHD, validity, and reliability in a Brazilian sample. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2019. [DOI: 10.1016/j.jpedp.2018.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
60
|
Costa DS, de Paula JJ, Malloy-Diniz LF, Romano-Silva MA, Miranda DM. Parent SNAP-IV rating of attention-deficit/hyperactivity disorder: accuracy in a clinical sample of ADHD, validity, and reliability in a Brazilian sample. J Pediatr (Rio J) 2019; 95:736-743. [PMID: 30236592 DOI: 10.1016/j.jped.2018.06.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/14/2018] [Accepted: 06/24/2018] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the psychometric properties of the short or multimodal treatment study version of the Swanson, Nolan, and Pelham, Version IV (SNAP-IV) scale, which measures attention-deficit/hyperactivity disorder and oppositional defiant disorder symptoms. METHODS Participants were 765 parents of children from 4 to 16 years old (641 non-attention-deficit/hyperactivity disorder and 124 attention-deficit/hyperactivity disorder children) from Belo Horizonte, Brazil, who reported sociodemographic characteristics and answered the SNAP-IV. Parents of the clinical sample also underwent the K-SADS-PL interview. RESULTS Age was significantly associated with SNAP-IV hyperactivity-impulsivity problems (r=-0.14), but not with inattention or oppositional defiant disorder. Sex was a significant influence on attention-deficit/hyperactivity disorder and oppositional defiant disorder severity (all p<0.001), with boys showing higher scores in the full sample, but not within the attention-deficit/hyperactivity disorder group. Exploratory and confirmatory factor analysis supports a three-factor structure of the SNAP-IV scale. Moderate-to-strong correlations were found between SNAP-IV and K-SADS-PL measures. All SNAP-IV scales showed very high internal consistency coefficients (all above 0.91). SNAP-IV inattention scores were the most predictive of attention-deficit/hyperactivity disorder diagnosis (AUC: 0.877 for the averaging rating method and the raw sum method, and 0.874 for the symptom presence/absence method). CONCLUSION The parent SNAP-IV showed good psychometric properties in a Brazilian school and clinical sample.
Collapse
Affiliation(s)
- Danielle S Costa
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Medicina Molecular, Belo Horizonte, MG, Brazil
| | - Jonas Jardim de Paula
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Medicina Molecular, Belo Horizonte, MG, Brazil; Faculdade de Ciências Médicas de Minas Gerais, Departamento de Psicologia, Belo Horizonte, MG, Brazil.
| | - Leandro F Malloy-Diniz
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Medicina Molecular, Belo Horizonte, MG, Brazil; Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Saúde Mental, Belo Horizonte, MG, Brazil
| | - Marco A Romano-Silva
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Medicina Molecular, Belo Horizonte, MG, Brazil; Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Saúde Mental, Belo Horizonte, MG, Brazil
| | - Débora M Miranda
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Medicina Molecular, Belo Horizonte, MG, Brazil; Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Pediatria, Belo Horizonte, MG, Brazil
| |
Collapse
|
61
|
Integrated Analysis of microRNA and mRNA Expression Profiles: An Attempt to Disentangle the Complex Interaction Network in Attention Deficit Hyperactivity Disorder. Brain Sci 2019; 9:brainsci9100288. [PMID: 31652596 PMCID: PMC6826944 DOI: 10.3390/brainsci9100288] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/16/2019] [Accepted: 10/20/2019] [Indexed: 12/11/2022] Open
Abstract
Attention Deficit Hyperactivity Disorder (ADHD) is a childhood-onset neurodevelopmental disorder, whose etiology and pathogenesis are still largely unknown. In order to uncover novel regulatory networks and molecular pathways possibly related to ADHD, we performed an integrated miRNA and mRNA expression profiling analysis in peripheral blood samples of children with ADHD and age-matched typically developing (TD) children. The expression levels of 13 miRNAs were evaluated with microfluidic qPCR, and differentially expressed (DE) mRNAs were detected on an Illumina HiSeq 2500 genome analyzer. The miRNA targetome was identified using an integrated approach of validated and predicted interaction data extracted from seven different bioinformatic tools. Gene Ontology (GO) and pathway enrichment analyses were carried out. Results showed that six miRNAs (miR-652-3p, miR-942-5p, let-7b-5p, miR-181a-5p, miR-320a, and miR-148b-3p) and 560 genes were significantly DE in children with ADHD compared to TD subjects. After correction for multiple testing, only three miRNAs (miR-652-3p, miR-148b-3p, and miR-942-5p) remained significant. Genes known to be associated with ADHD (e.g., B4GALT2, SLC6A9 TLE1, ANK3, TRIO, TAF1, and SYNE1) were confirmed to be significantly DE in our study. Integrated miRNA and mRNA expression data identified critical key hubs involved in ADHD. Finally, the GO and pathway enrichment analyses of all DE genes showed their deep involvement in immune functions, reinforcing the hypothesis that an immune imbalance might contribute to the ADHD etiology. Despite the relatively small sample size, in this study we were able to build a complex miRNA-target interaction network in children with ADHD that might help in deciphering the disease pathogenesis. Validation in larger samples should be performed in order to possibly suggest novel therapeutic strategies for treating this complex disease.
Collapse
|
62
|
Jin J, Liu L, Chen W, Gao Q, Li H, Wang Y, Qian Q. The Implicated Roles of Cell Adhesion Molecule 1 ( CADM1) Gene and Altered Prefrontal Neuronal Activity in Attention-Deficit/Hyperactivity Disorder: A "Gene-Brain-Behavior Relationship"? Front Genet 2019; 10:882. [PMID: 31616473 PMCID: PMC6775240 DOI: 10.3389/fgene.2019.00882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 08/21/2019] [Indexed: 12/16/2022] Open
Abstract
Background: Genes related to cell adhesion pathway have been implicated in the genetic architecture of attention-deficit/hyperactivity disorder (ADHD). Cell adhesion molecule 1, encoded by CADM1 gene, is a protein which facilitates cell adhesion, highly expressed in the human prefrontal lobe. This study aimed to evaluate the association of CADM1 genotype with ADHD, executive function, and regional brain functions. Methods: The genotype data of 10-tag single nucleotide polymorphisms of CADM1 for 1,040 children and adolescents with ADHD and 963 controls were used for case–control association analyses. Stroop color–word interference test, Rey–Osterrieth complex figure test, and trail making test were conducted to assess “inhibition,” “working memory,” and “set-shifting,” respectively. A subsample (35 ADHD versus 56 controls) participated in the nested imaging genetic study. Resting-state functional magnetic resonance images were acquired, and the mean amplitude of low-frequency fluctuations (mALFF) were captured. Results: Nominal significant genotypic effect of rs10891819 in “ADHD-alone” subgroup was detected (P = 0.008) with TT genotype as protective. The results did not survive multiple testing correction. No direct genetic effect was found for performance on executive function tasks. In the imaging genetic study for the “ADHD-whole” sample, rs10891819 genotype was significantly associated with altered mALFF in the right superior frontal gyrus (rSFG, peak t = 3.85, corrected P < 0.05). Specifically, the mALFFs in T-allele carriers were consistently higher than GG carriers in ADHD and control groups. Endophenotypic correlation analyses indicated a significant negative correlation between “word interference time” in Stroop (shorter “word interference time” indexing better inhibitory function) and mALFF in the rSFG (r = -0.29, P = 0.006). Finally, mediation analysis confirmed significant indirect effects from “rs10891819 genotype (T-allele carriers)” via “mALFF (rSFG)” to “inhibition (“word interference time”)” (Sobelz = -2.47; B = -2.61, 95% confidence interval -0.48 to -4.72; P = 0.009). Conclusions: Our study offered preliminary evidence to implicate the roles of CADM1 in relation to prefrontal brain activities, inhibition function, and ADHD, indicating a potential “gene–brain–behavior” relationship of the CADM1 gene. Future studies with larger samples may specifically test these hypotheses generated by our exploratory findings.
Collapse
Affiliation(s)
- Jiali Jin
- Department of Child Psychiatry, Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.,National Clinical Research Center for Mental Disorders & the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Lu Liu
- Department of Child Psychiatry, Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.,National Clinical Research Center for Mental Disorders & the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Wai Chen
- Centre & Discipline of Child and Adolescent Psychiatry, and Psychotherapy, School of Medicine, Division of Paediatrics and Child Health & Division of Psychiatry and Clinical Neurosciences, The University of Western Australia, Perth, WA, Australia.,Complex Attention and Hyperactivity Disorders Service (CAHDS), Specialised Child and Adolescent Mental Health Services of Health in Western Australia, Perth, WA, Australia
| | - Qian Gao
- Department of Child Psychiatry, Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.,National Clinical Research Center for Mental Disorders & the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Haimei Li
- Department of Child Psychiatry, Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.,National Clinical Research Center for Mental Disorders & the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Yufeng Wang
- Department of Child Psychiatry, Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.,National Clinical Research Center for Mental Disorders & the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Qiujin Qian
- Department of Child Psychiatry, Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.,National Clinical Research Center for Mental Disorders & the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| |
Collapse
|
63
|
Pineau G, Villemonteix T, Slama H, Kavec M, Balériaux D, Metens T, Baijot S, Mary A, Ramoz N, Gorwood P, Peigneux P, Massat I. Dopamine transporter genotype modulates brain activity during a working memory task in children with ADHD. RESEARCH IN DEVELOPMENTAL DISABILITIES 2019; 92:103430. [PMID: 31306870 DOI: 10.1016/j.ridd.2019.103430] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/04/2019] [Accepted: 06/23/2019] [Indexed: 06/10/2023]
Abstract
Dopamine active transporter gene (DAT1) is a candidate gene associated with attention-deficit/hyperactivity disorder (ADHD). The DAT1 variable number tandem repeat (VNTR)-3' polymorphism is functional and 9R carriers have been shown to produce more DAT than 10R homozygotes. We used functional magnetic resonance imaging (fMRI) to investigate the effects of this polymorphism on the neural substrates of working memory (WM) in a small but selected population of children with ADHD, naïve of any psychotropic treatment and without comorbidity. MRI and genotype data were obtained for 36 children (mean age: 10,36 +/- 1,49 years) with combined-type ADHD (9R n = 15) and 25 typically developing children (TDC) (mean age: 9,55 +/- 1,25 years) (9R n = 12). WM performance was similar between conditions. We found a cross-over interaction effect between gene (9R vs. 10R) and diagnosis (TDC vs. ADHD) in the orbito-frontal gyrus, cerebellum and inferior temporal lobe. In these areas, WM-related activity was higher for 9R carriers in ADHD subjects and lower in TDC. In ADHD children only, 10R homozygotes exhibited higher WM-related activity than 9R carriers in a network encompassing the parietal and the temporal lobes, the ventral visual cortex, the orbito-frontal gyrus and the head of the caudate nucleus. There was no significant results in TDC group. Our preliminary findings suggest that DAT1 VNTR polymorphism can modulate WM-related brain activity ADHD children.
Collapse
Affiliation(s)
- Guillaume Pineau
- GHU Paris Psychiatrie and Neurosciences (CMME, Hôpital Sainte-Anne), 1 rue Cabanis, 75014 Paris, France; INSERM U1266 (Team 1, Institute of Psychiatry and Neurosciences of Paris), Paris, France.
| | - Thomas Villemonteix
- Psychopathology and Neuropsychology Lab, Paris 8 University, 2 Rue de la Liberté, 93526 Saint-Denis, France
| | - Hichem Slama
- UR2NF - Neuropsychology and Functional Neuroimaging Research Unit at CRCN - Centre de Recherche Cognition et Neurosciences and UNI - ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Belgium; UNESCOG - Research Unit in Cognitive Neurosciences, at CRCN - Centre de Recherche Cognition et Neurosciences and UNI - ULB Neurosciences Institute, ULB, Belgium; Department of Clinical and Cognitive Neuropsychology, Erasme Hospital, Belgium
| | - Martin Kavec
- Department of Radiology, Clinics of Magnetic Resonance, Erasme Hospital, 808 Lennik street, CP601, 1070 Brussels, Belgium; UNI - ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Belgium
| | - Danielle Balériaux
- Department of Radiology, Clinics of Magnetic Resonance, Erasme Hospital, 808 Lennik street, CP601, 1070 Brussels, Belgium; UNI - ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Belgium
| | - Thierry Metens
- Department of Radiology, Clinics of Magnetic Resonance, Erasme Hospital, 808 Lennik street, CP601, 1070 Brussels, Belgium; UNI - ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Belgium
| | - Simon Baijot
- UR2NF - Neuropsychology and Functional Neuroimaging Research Unit at CRCN - Centre de Recherche Cognition et Neurosciences and UNI - ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Belgium; UNESCOG - Research Unit in Cognitive Neurosciences, at CRCN - Centre de Recherche Cognition et Neurosciences and UNI - ULB Neurosciences Institute, ULB, Belgium
| | - Alison Mary
- UR2NF - Neuropsychology and Functional Neuroimaging Research Unit at CRCN - Centre de Recherche Cognition et Neurosciences and UNI - ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Belgium; National Fund of Scientific Research, Belgium
| | - Nicolas Ramoz
- INSERM U1266 (Team 1, Institute of Psychiatry and Neurosciences of Paris), Paris, France
| | - Philip Gorwood
- GHU Paris Psychiatrie and Neurosciences (CMME, Hôpital Sainte-Anne), 1 rue Cabanis, 75014 Paris, France; INSERM U1266 (Team 1, Institute of Psychiatry and Neurosciences of Paris), Paris, France
| | - Philippe Peigneux
- UR2NF - Neuropsychology and Functional Neuroimaging Research Unit at CRCN - Centre de Recherche Cognition et Neurosciences and UNI - ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Belgium
| | - Isabelle Massat
- UR2NF - Neuropsychology and Functional Neuroimaging Research Unit at CRCN - Centre de Recherche Cognition et Neurosciences and UNI - ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Belgium; National Fund of Scientific Research, Belgium; Laboratory of Experimental Neurology, ULB, Belgium
| |
Collapse
|
64
|
Heim CM, Entringer S, Buss C. Translating basic research knowledge on the biological embedding of early-life stress into novel approaches for the developmental programming of lifelong health. Psychoneuroendocrinology 2019; 105:123-137. [PMID: 30578047 PMCID: PMC6561839 DOI: 10.1016/j.psyneuen.2018.12.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/22/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022]
Abstract
This review integrates scientific knowledge obtained over the past few decades on the biological mechanisms that contribute to the profound association between exposure to early adversity, including childhood trauma and prenatal stress, and the lifelong elevated risk to develop a broad range of diseases. We further discuss insights into gene-environment interactions moderating the association between early adversity and disease manifestation and we discuss the role of epigenetic and other molecular processes in the biological embedding of early adversity. Based on these findings, we propose potential mechanisms that may contribute to the intergenerational transmission of risk related to early adversity from the mother to the fetus. Finally, we argue that basic research knowledge on the biological embedding of early adversity must now be translated into novel intervention strategies that are mechanism-driven and sensitive to developmental timing. Indeed, to date, there are no diagnostic biomarkers of risk or mechanism-informed interventions that we can offer to victims of early adversity in order to efficiently prevent or reverse adverse health outcomes. Such translational efforts can be expected to have significant impact on both clinical practice and the public health system, and will promote precision medicine in pediatrics and across the lifespan.
Collapse
Affiliation(s)
- Christine M. Heim
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany,Department of Biobehavioral Health, College of Health & Human Development, The Pennsylvania State University, University Park, PA, USA,Corresponding authors at: Institute of Medical Psychology, Charité Universitätsmedizin Berlin, Luisenstr. 57, 10117 Berlin, Germany., (C.M. Heim), (S. Entringer), (C. Buss)
| | - Sonja Entringer
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany; Development, Health, and Disease Research Program, University of California Irvine, Orange, CA, USA.
| | - Claudia Buss
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany; Development, Health, and Disease Research Program, University of California Irvine, Orange, CA, USA.
| |
Collapse
|
65
|
Sekaninova N, Mestanik M, Mestanikova A, Hamrakova A, Tonhajzerova I. Novel approach to evaluate central autonomic regulation in attention deficit/hyperactivity disorder (ADHD). Physiol Res 2019; 68:531-545. [PMID: 31177787 DOI: 10.33549/physiolres.934160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Attention deficit/hyperactivity disorder (ADHD) is one of the most commonly diagnosed developmental disorders in childhood characterized by hyperactivity, impulsivity and inattention. ADHD manifests in the child's development by deficits in cognitive, executive and perceptor-motor functions, emotional regulation and social adaptation. Although the exact cause has not yet been known, the crucial role in the development of this disease plays the interaction of genetic, neurobiological and epigenetic factors. According to current knowledge, ADHD is defined as a biological dysfunction of central nervous system with genetically or organically defined deficits in noradrenergic and dopaminergic neurotransmission associated with structural abnormalities, especially in prefronto-striatal regions. In this context, a significant part of the difficulties could be due to a faulty control of fronto-striato-thalamo-cortical circuits important for attention, arousal and executive functions. Moreover, ADHD is associated with abnormal autonomic regulation. Specifically, reduced cardiac-linked parasympathetic activity associated with relative sympathetic dominance indexed by low heart rate variability can represent a noninvasive marker for prefrontal hypoactivity. However, the mechanisms underlying altered autonomic regulation in ADHD are still unknown. In this aspect, the evaluation of central autonomic regulation by noninvasive methods, namely pupillometry and eye-tracking, may provide novel information for better understanding of the neurobiological pathomechanisms leading to ADHD.
Collapse
Affiliation(s)
- N Sekaninova
- Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic.
| | | | | | | | | |
Collapse
|
66
|
Huang X, Zhang Q, Gu X, Hou Y, Wang M, Chen X, Wu J. LPHN3 gene variations and susceptibility to ADHD in Chinese Han population: a two-stage case-control association study and gene-environment interactions. Eur Child Adolesc Psychiatry 2019; 28:861-873. [PMID: 30406846 DOI: 10.1007/s00787-018-1251-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 10/31/2018] [Indexed: 12/18/2022]
Abstract
Polymorphisms in latrophilin 3 (LPHN3) were recently reported to be associated with attention-deficit/hyperactivity disorder (ADHD), and subsequently other researchers tried to replicate the findings in different populations. This study was aimed to confirm the role of the LPHN3 in ADHD and explore the potential interactions with environmental risk factors in Chinese Han population. We examined the association of LPHN3 with ADHD in a population of 473 ADHD children and 585 controls. As a supplement of ADHD diagnosis, Conners Parent Symptom Questionnaire (PSQ) was used to evaluate ADHD symptoms. Blood lead levels (BLLs) were measured by atomic absorption spectrophotometry and other potential environmental risk factors were determined via a questionnaire filled out by the parents. Finally, after validation in an independent sample (284 cases and 390 controls), we observed significant associations between LPHN3 variants rs1868790 and ADHD risk in combined stage within codominant model [TA/AA: OR (95% CI) = 1.636 (1.325-2.021)], dominant model [OR (95% CI) = 1.573 (1.288-1.922)], and additive model [OR (95% CI) = 1.535 (1.266-1.862)]. Furthermore, rs1868790 significantly interacted with BLLs and maternal stress to modify ADHD susceptibility (P < 0.05), and rs1868790 was found to be related with ADHD symptoms (P < 0.05). Expression quantitative trait loci analysis further indicated that rs1868790 took part in the regulation of LPHN3 gene expression. As the first study to comprehensively explore the role of LPHN3 in ADHD in Chinese children, our research suggests that LPHN3 gene has a significant effect on the ADHD in a Chinese population.
Collapse
Affiliation(s)
- Xin Huang
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qi Zhang
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xue Gu
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yuwei Hou
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Min Wang
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xinzhen Chen
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jing Wu
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China. .,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
67
|
Huang X, Wang M, Zhang Q, Chen X, Wu J. The role of glutamate receptors in attention-deficit/hyperactivity disorder: From physiology to disease. Am J Med Genet B Neuropsychiatr Genet 2019; 180:272-286. [PMID: 30953404 DOI: 10.1002/ajmg.b.32726] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/12/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022]
Abstract
Attention-deficit hyperactivity disorder (ADHD) is the most common psychiatric disorder in children and adolescents, which is characterized by behavioral problems such as attention deficit, hyperactivity, and impulsivity. As the receptors of the major excitatory neurotransmitter in the mammalian central nervous system (CNS), glutamate receptors (GluRs) are strongly linked to normal brain functioning and pathological processes. Extensive investigations have been made about the structure, function, and regulation of GluR family, describing evidences that support the disruption of these mechanisms in mental disorders, including ADHD. In this review, we briefly described the family and function of GluRs in the CNS, and discussed what is recently known about the role of GluRs in ADHD, that including GluR genes, animal models, and the treatment, which would help us further elucidate the etiology of ADHD.
Collapse
Affiliation(s)
- Xin Huang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinzhen Chen
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
68
|
Genetic risk factors and gene–environment interactions in adult and childhood attention-deficit/hyperactivity disorder. Psychiatr Genet 2019; 29:63-78. [DOI: 10.1097/ypg.0000000000000220] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
69
|
Huang X, Zhang Q, Chen X, Gu X, Wang M, Wu J. A functional variant in SLC1A3 influences ADHD risk by disrupting a hsa-miR-3171 binding site: A two-stage association study. GENES BRAIN AND BEHAVIOR 2019; 18:e12574. [PMID: 30953407 DOI: 10.1111/gbb.12574] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/06/2019] [Accepted: 04/03/2019] [Indexed: 12/14/2022]
Abstract
Attention-deficit hyperactivity disorder (ADHD) is one of the most common neuropsychiatric disorders in children and adolescents with high heritability. Evidence is accumulating that SLC1A3 may play a role in ADHD etiology. Therefore, a two-stage case-control study was conducted on 752 cases and 774 controls to explore the role of SLC1A3 in ADHD. Bioinformatic annotations and functional experiments were applied to reveal the potential biological mechanisms. Finally, SLC1A3 rs1049522 showed significant association with ADHD risk in two stages with CA genotype vs AA genotype, odds ratio (OR) = 0.694 (95% confidence interval, CI = 0.570-0.844) and dominant model, OR = 0.749 (95% CI = 0.621-0.904) in the combined stage. Besides, rs1049522 was found to be related to ADHD hyperactive/impulsive symptom, and rs1049522-C showed increased SLC1A3 mRNA expression in the cerebellar cortex. Dual-luciferase reporter assay further indicated that rs1049522-C allele enhanced SLC1A3 expression by disrupting the hsa-miR-3171 binding site. In conclusion, SLC1A3 variant rs1049522 was implicated in ADHD susceptibility in a Chinese Han population probably by enhancing the SLC1A3 expression in a miRNA-mediated manner.
Collapse
Affiliation(s)
- Xin Huang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qi Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xinzhen Chen
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xue Gu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Min Wang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jing Wu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
70
|
Fontana BD, Franscescon F, Rosemberg DB, Norton WH, Kalueff AV, Parker MO. Zebrafish models for attention deficit hyperactivity disorder (ADHD). Neurosci Biobehav Rev 2019; 100:9-18. [DOI: 10.1016/j.neubiorev.2019.02.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 01/23/2023]
|
71
|
Böhm S, Curran EA, Kenny LC, O'Keeffe GW, Murray D, Khashan AS. The Effect of Hypertensive Disorders of Pregnancy on the Risk of ADHD in the Offspring. J Atten Disord 2019; 23:692-701. [PMID: 28162026 DOI: 10.1177/1087054717690230] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Evidence suggests that perinatal factors may contribute to the development of ADHD. Our objective was to examine the association between hypertensive disorders of pregnancy (HDP) and ADHD, and behavioral difficulties among 7-year-old children. METHOD The study cohort consisted of 13,192 children (weighted = 13,500) who participated in the Millennium Cohort Study (MCS) at age 7. HDP (raised blood pressure, preeclampsia, eclampsia, and toxemia) were reported by mothers 9 months postdelivery. ADHD was reported by parents at age 7 years. Weighted logistic regression models were used to assess the association. RESULTS In all, 1,069 (7.9%) women reported HDP and 166 (1.2%) children had an ADHD diagnosis. There was a significant association between HDP and ADHD (adjusted odds ratio [OR] = 1.78, 95% confidence interval [CI] = [1.03, 3.07]). CONCLUSION These findings suggest that HDP is associated with an increased risk of ADHD. It is important to confirm this in larger cohorts and to understand the biological basis of this association.
Collapse
|
72
|
Morgan JE, Lee SS, Loo SK. Fluid Reasoning Mediates the Association of Birth Weight With ADHD Symptoms in Youth From Multiplex Families With ADHD. J Atten Disord 2019; 23:682-691. [PMID: 27658748 PMCID: PMC5360552 DOI: 10.1177/1087054716670006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE We tested mediation of birth weight and ADHD symptoms by multiple biologically plausible neurocognitive functions and evaluated familiality of observed indirect effects. METHOD 647 youth from 284 multiplex families with ADHD completed the Arithmetic, Digit Span, Vocabulary, and Block Design subtests of the Wechsler Intelligence Scale for Children (WISC). Multiple mediation tested WISC subtests as mediators of birth weight and multi-informant ADHD symptoms. Familiality of indirect effects was estimated via moderated mediation comparing conditional indirect effects across siblings concordant and discordant for ADHD. RESULTS Controlling for IQ and demographic factors, Arithmetic uniquely mediated birth weight and ADHD symptoms. Conditional indirect effects through Arithmetic did not differ across ADHD concordant and discordant siblings. CONCLUSION These cross-sectional findings support previous prospective longitudinal research implicating Arithmetic (i.e., fluid reasoning) as a preliminary causal mediator of birth weight and ADHD symptoms, and suggest that this pathway is independent of genetic influences on ADHD.
Collapse
|
73
|
Changes in serum miRNA-let-7 level in children with attention deficit hyperactivity disorder treated by repetitive transcranial magnetic stimulation or atomoxetine: An exploratory trial. Psychiatry Res 2019; 274:189-194. [PMID: 30807970 DOI: 10.1016/j.psychres.2019.02.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
We aimed to investigate whether microRNA-let-7d (miRNA-let-7d) and miRNA-107 may serve as diagnostic and therapeutic biomarkers of attention deficit hyperactivity disorder (ADHD). The relative expression level of miRNA-let-7d and miRNA-107 in patients with ADHD and in a healthy control group was detected by real-time polymerase chain reaction. The blood samples were collected at 6 weeks after repetitive transcranial magnetic stimulation (rTMS) or atomoxetine (ATX) in ADHD patients, and the relative expression levels of the two miRNAs before and after treatments were compared. There were significant differences in the expression level of miRNA-let-7d between ADHD patients and healthy children, as well as before and after rTMS or ATX treatment in ADHD patients. However, the expression of miRNA-107 showed no significant difference between ADHD patients and healthy children or before and after rTMS (or ATX treatment). These results suggest that serum miRNA-let-7d may serve as a potential diagnostic and therapeutic biomarker for children with ADHD.
Collapse
|
74
|
Mustafin RN, Enikeeva RF, Malykh SB, Valinurov RG, Khusnutdinova EK. [Genetics and epigenetics of attention deficit hyperactivity disorder]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 118:106-110. [PMID: 30335081 DOI: 10.17116/jnevro2018118091106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Molecular genetic studies of attention deficit hyperactivity disorder (ADHD) have demonstrated the involvement of multiple genes in the etiology of ADHD. A polygenic hypothesis of the etiopathogenesis was formulated without clear knowledge of common mechanisms of ADHD development. Twin, family and adoption studies have established the heritability of 70-80% for ADHD. Association studies have shown the relationship between ADHD and genes of dopaminergic (DRD4, DRD5, SLC6A3), serotoninergic (HTR1B, 5-HTTLPR), glutamatergic (mGluR, NDRG2) systems, metabolic pathways (SLC2A3, SLC6A4, CDH13, CFOD1, GFOD1), membrane proteins (KChIP1, ITGA1, SNAP-25) as well as tumour-suppressor (NDRG2, NF1) and cytokine genes. The marked comorbidity of ADHD with other psychiatric disorders and shared genetic risk factors were determined. Studies of a role of copy number variations (CNVs) provided more promising evidence that suggested the possible involvement of retroelements as the unifying factors of disease etiopathogenesis. Transposons, which are sensitive to stress, may cause CNVs and are key regulators of brain development and functioning. The dysregulation of transposons is thought to be important in changes in tuning of gene regulatory pathways and epigenetic regulation of neurons in ADHD that may be a common principle underlying the heterogeneous nature of ADHD. Research on noncoding RNAs will help to confirm the hypothesis and develop diagnostic algorithms of examination of ADHD patients as an important step in the implementation of personalized medicine in psychiatry.
Collapse
Affiliation(s)
| | - R F Enikeeva
- Bashkir State University, Ufa, Russia; Institute of Biochemistry and Genetics of the Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia
| | - S B Malykh
- Psychological Institute of Russian Academy of Education, Moscow, Russia
| | | | - E K Khusnutdinova
- Bashkir State University, Ufa, Russia; Institute of Biochemistry and Genetics of the Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia
| |
Collapse
|
75
|
Martinhago F, Lavagnino NJ, Folguera G, Caponi S. Risk factors and genetic bases: the case of attention deficit hyperactivity disorder. Salud Colect 2019; 15:e1952. [PMID: 31664338 DOI: 10.18294/sc.2019.1952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/21/2018] [Indexed: 11/24/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is considered to be the most frequent mental disorder in childhood. Although its diagnosis in the most utilized handbook of psychiatry in the world today - the Diagnostic and statistical handbook of mental disorders (DSM-5) - is based on behaviors of inattention, hyperactivity and impulsivity, numerous attempts to describe the biological bases of the disorder can be found, to be used for and also as risk markers. In this paper, we will critically analyze the validity of studies associated with the search for genetic markers of ADHD. First, a characterization of ADHD by the DSM-5 handbook is presented. Subsequently, the link between ADHD, risk factors and genetic markers is developed. Finally, some conclusions are presented which highlight simplifications and omissions that could have significant consequences.
Collapse
Affiliation(s)
- Fernanda Martinhago
- Doctora en Ciencias Humanas. Doctora en Antropología y Comunicación. Posdoctoranda, Programa de Pós-Graduação Interdisciplinar em Ciências Humanas, Universidade Federal de Santa Catarina. Becaria, Coordenação de Aperfeiçoamento de Pessoal de Nivel Superior, Programa Nacional de Pós-Doutorado; Florianópolis, Brasil.
| | - Nicolás José Lavagnino
- Licenciado en Ciencias Biológicas. Licenciado en Filosofía. Doctor en Ciencias Biológicas. Investigador Asistente, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Guillermo Folguera
- Licenciado en Ciencias Biológicas. Licenciado en Filosofía. Doctor en Ciencias Biológicas. Profesor, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Investigador Adjunto, Consejo Nacional de Investigaciones Científicas y Técnicas. Ciudad Autónoma de Buenos Aires, Argentina.
| | - Sandra Caponi
- Doctora en Filosofía. Profesora titular, Departamento de Sociologia e Ciência Política, Universidade Federal de Santa Catarina. Becaria Senior, Coordenação de Aperfeiçoamento de Pessoal de Nivel Superior. Florianópolis, Brasil.
| |
Collapse
|
76
|
Pharmacological treatment of attention-deficit hyperactivity disorder comorbid with an anxiety disorder: a systematic review. Int Clin Psychopharmacol 2019; 34:57-64. [PMID: 30422834 DOI: 10.1097/yic.0000000000000243] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The purpose of this study was to conduct a systematic review of the pharmacological options available to treat patients diagnosed with attention-deficit hyperactivity disorder and anxiety disorder, for generating evidence on the safest, most-effective and tolerable pharmacotherapy. To this end, a systematic search was performed in three electronic databases (Medline, Scopus and Directory of Open Access Journals; December 2017). Randomized, double-blind, parallel-design clinical trials evaluating the efficacy, safety or tolerability of therapies for attention-deficit hyperactivity disorder and anxiety disorder in children and adolescents or adults were considered. A total of 1960 articles were retrieved from the databases, of which five studies were included in the qualitative synthesis. Two of these studies evaluated the drug atomoxetine, another study evaluated desipramine, and the remaining two studies evaluated methylphenidate, with fluvoxamine being associated with methylphenidate in one of the trials. Owing to the high heterogeneity among studies, it was not possible to combine data for meta-analyses. Although only few studies have been evaluated in this systematic review, the results point to a more significant benefit of atomoxetine. This is probably because this drug was studied in a wider age range and evaluated by more specific scales for both disorders. To further strengthen this evidence, randomized, controlled and multicenter clinical trials with larger sample sizes should be conducted.
Collapse
|
77
|
Polygenic Risk and Neural Substrates of Attention-Deficit/Hyperactivity Disorder Symptoms in Youths With a History of Mild Traumatic Brain Injury. Biol Psychiatry 2019; 85:408-416. [PMID: 30119875 PMCID: PMC6330150 DOI: 10.1016/j.biopsych.2018.06.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/12/2018] [Accepted: 06/28/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Attention-deficit/hyperactivity disorder (ADHD) is a major sequela of traumatic brain injury (TBI) in youths. The objective of this study was to examine whether ADHD symptoms are differentially associated with genetic risk and brain structure in youths with and without a history of TBI. METHODS Medical history, ADHD symptoms, genetic data, and neuroimaging data were obtained from a community sample of youths. ADHD symptom severity was compared between those with and without TBI (TBI n = 418, no TBI n = 3193). The relationship of TBI history, genetic vulnerability, brain structure, and ADHD symptoms was examined by assessing 1) ADHD polygenic score (discovery sample ADHD n = 19,099, control sample n = 34,194), 2) basal ganglia volumes, and 3) fractional anisotropy in the corpus callosum and corona radiata. RESULTS Youths with TBI reported greater ADHD symptom severity compared with those without TBI. Polygenic score was positively associated with ADHD symptoms in youths without TBI but not in youths with TBI. The negative association between the caudate volume and ADHD symptoms was not moderated by a history of TBI. However, the relationship between ADHD symptoms and structure of the genu of the corpus callosum was negative in youths with TBI and positive in youths without TBI. CONCLUSIONS The identification of distinct ADHD etiology in youths with TBI provides neurobiological insight into the clinical heterogeneity in the disorder. Results indicate that genetic predisposition to ADHD does not increase the risk for ADHD symptoms associated with TBI. ADHD symptoms associated with TBI may be a result of a mechanical insult rather than neurodevelopmental factors.
Collapse
|
78
|
Findling RL, Adler LA, Spencer TJ, Goldman R, Hopkins SC, Koblan KS, Kent J, Hsu J, Loebel A. Dasotraline in Children with Attention-Deficit/Hyperactivity Disorder: A Six-Week, Placebo-Controlled, Fixed-Dose Trial. J Child Adolesc Psychopharmacol 2019; 29:80-89. [PMID: 30694697 DOI: 10.1089/cap.2018.0083] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Dasotraline is a potent inhibitor of presynaptic dopamine and norepinephrine reuptake with a pharmacokinetic profile characterized by slow absorption and a long elimination half-life. The aim of this study was to evaluate the efficacy and safety of dasotraline in children with attention-deficit/hyperactivity disorder (ADHD). METHODS Children aged 6-12 years with a Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5) diagnosis of ADHD were randomized to 6 weeks of double-blind once-daily treatment with dasotraline (2 or 4 mg) or placebo. The primary efficacy endpoint was change from baseline in the ADHD Rating Scale Version IV-Home Version (ADHD RS-IV HV) total score at week 6. RESULTS A total of 342 patients were randomized to dasotraline or placebo (mean age 9.1 years, 66.7% male). Treatment with dasotraline was associated with significant improvement at study endpoint in the ADHD RS-IV HV total score for the 4 mg/day dose versus placebo (-17.5 vs. -11.4; p < 0.001; effect size [ES], 0.48), but not for the 2 mg/day dose (-11.8 vs. -11.4; ns; ES, 0.03). A regression analysis confirmed a significant linear dose-response relationship for dasotraline. Significant improvement for dasotraline 4 mg/day dose versus placebo was also observed across the majority of secondary efficacy endpoints, including the Clinical Global Impression (CGI)-Severity score, the Conners Parent Rating Scale-Revised scale (CPRS-R) ADHD index score, and subscale measures of hyperactivity and inattentiveness. Discontinuation rates due to adverse events (AEs) were higher in the dasotraline 4 mg/day group (12.2%) compared with the 2 mg/day group (6.3%) and placebo (1.7%). The most frequent AEs associated with dasotraline were insomnia, decreased appetite, decreased weight, and irritability. Psychosis-related symptoms were reported as AEs by 7/219 patients treated with dasotraline in this study. There were no serious AEs or clinically meaningful changes in blood pressure or heart rate on dasotraline. CONCLUSION In this placebo-controlled study, treatment with dasotraline 4 mg/day significantly improved ADHD symptoms and behaviors, including attention and hyperactivity, in children aged 6-12 years. The most frequently reported AEs observed on dasotraline included insomnia, decreased appetite, decreased weight, and irritability.
Collapse
Affiliation(s)
- Robert L Findling
- 1 Kennedy Krieger Institute/Johns Hopkins University , Baltimore, Maryland
| | - Lenard A Adler
- 2 New York University Langone Medical Center , New York, New York
| | | | - Robert Goldman
- 4 Sunovion Pharmaceuticals, Inc. , Marlborough, Massachusetts and Fort Lee, New Jersey
| | - Seth C Hopkins
- 4 Sunovion Pharmaceuticals, Inc. , Marlborough, Massachusetts and Fort Lee, New Jersey
| | - Kenneth S Koblan
- 4 Sunovion Pharmaceuticals, Inc. , Marlborough, Massachusetts and Fort Lee, New Jersey
| | - Justine Kent
- 4 Sunovion Pharmaceuticals, Inc. , Marlborough, Massachusetts and Fort Lee, New Jersey
| | - Jay Hsu
- 4 Sunovion Pharmaceuticals, Inc. , Marlborough, Massachusetts and Fort Lee, New Jersey
| | - Antony Loebel
- 4 Sunovion Pharmaceuticals, Inc. , Marlborough, Massachusetts and Fort Lee, New Jersey
| |
Collapse
|
79
|
Dalla Vecchia E, Mortimer N, Palladino VS, Kittel-Schneider S, Lesch KP, Reif A, Schenck A, Norton WH. Cross-species models of attention-deficit/hyperactivity disorder and autism spectrum disorder: lessons from CNTNAP2, ADGRL3, and PARK2. Psychiatr Genet 2019; 29:1-17. [PMID: 30376466 PMCID: PMC7654943 DOI: 10.1097/ypg.0000000000000211] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 12/12/2022]
Abstract
Animal and cellular models are essential tools for all areas of biological research including neuroscience. Model systems can also be used to investigate the pathophysiology of psychiatric disorders such as attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). In this review, we provide a summary of animal and cellular models for three genes linked to ADHD and ASD in human patients - CNTNAP2, ADGRL3, and PARK2. We also highlight the strengths and weaknesses of each model system. By bringing together behavioral and neurobiological data, we demonstrate how a cross-species approach can provide integrated insights into gene function and the pathogenesis of ADHD and ASD. The knowledge gained from transgenic models will be essential to discover and validate new treatment targets for these disorders.
Collapse
Affiliation(s)
- Elisa Dalla Vecchia
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Niall Mortimer
- Division of Molecular Psychiatry, Centre of Mental Health, University of Wuerzburg, Wuerzburg
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona
- Department of Psychiatry, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Viola S. Palladino
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt Am Main, Germany
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt Am Main, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Centre of Mental Health, University of Wuerzburg, Wuerzburg
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt Am Main, Germany
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - William H.J. Norton
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| |
Collapse
|
80
|
Chen X, Wang M, Zhang Q, Hou Y, Huang X, Li S, Wu J. Stress response genes associated with attention deficit hyperactivity disorder: A case-control study in Chinese children. Behav Brain Res 2019; 363:126-134. [PMID: 30707907 DOI: 10.1016/j.bbr.2019.01.051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/26/2019] [Accepted: 01/26/2019] [Indexed: 01/12/2023]
Abstract
To explore the associations between stress response genes and attention deficit hyperactivity disorder (ADHD) in children, we conducted a case-control study consisting of 406 newly diagnosed ADHD cases and 432 controls in Wuhan, China. We genotyped the candidate genes, nuclear receptor subfamily 3 group C member 1(NR3C1) and solute carrier family 6 member 4(SLC6A4), using the Sequenom MassARRAY technology. After correction by Bonferroni (α' = 0.05/6 = 0.008), the rs6191 SNP was found to be associated with a reduced risk of ADHD in the dominant model (OR = 0.564, 95% CI = 0.389-0.819, P = 0.003) while the rs25531 SNP was associated with an increased risk of ADHD in the multiplicative model (OR = 1.380, 95% CI = 1.111-1.714, P = 0.004). Additionally, both the rs6191 and rs25531 SNPs were significantly associated with the attention deficit factor (P = 0.006, P = 0.003, respectively) but not with the hyperactivity/impulsivity factor in the Swanson, Nolan and Pelham-IV Questionnaire (SNAP-IV) scale. Furthermore, we found that these two SNPs were significantly associated with pure ADHD, and not affected by the comorbidities (P = 0.001, P = 0.007, respectively). Besides, there was an interaction between these two SNPs. This study demonstrated the role of NR3C1 and SLC6A4 polymorphisms in ADHD, yet independent replication of the findings of this study in multi-center and multi-stage studies with large samples is warranted in the future.
Collapse
Affiliation(s)
- Xinzhen Chen
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuwei Hou
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Huang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanyawen Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
81
|
Kappel DB, Schuch JB, Rovaris DL, da Silva BS, Müller D, Breda V, Teche SP, S Riesgo R, Schüler-Faccini L, Rohde LA, Grevet EH, Bau CHD. ADGRL3 rs6551665 as a Common Vulnerability Factor Underlying Attention-Deficit/Hyperactivity Disorder and Autism Spectrum Disorder. Neuromolecular Med 2019; 21:60-67. [PMID: 30652248 DOI: 10.1007/s12017-019-08525-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 01/10/2019] [Indexed: 12/27/2022]
Abstract
Neurodevelopmental disorders are prevalent, frequently occur in comorbidity and share substantial genetic correlation. Previous evidence has suggested a role for the ADGRL3 gene in Attention-Deficit/Hyperactivity Disorder (ADHD) susceptibility in several samples. Considering ADGRL3 functionality in central nervous system development and its previous association with neurodevelopmental disorders, we aimed to assess ADGRL3 influence in early-onset ADHD (before 7 years of age) and Autism Spectrum Disorder (ASD). The sample comprises 187 men diagnosed with early-onset ADHD, 135 boys diagnosed with ASD and 468 male blood donors. We tested the association of an ADGRL3 variant (rs6551665) with both early-onset ADHD and ASD susceptibility. We observed significant associations between ADGRL3-rs6551665 on ADHD and ASD susceptibilities; we found that G-carriers were at increased risk of ADHD and ASD, in accordance with previous studies. The overall evidence from the literature, corroborated by our results, suggests that ADGRL3 might be involved in brain development, and genetic modifications related to it might be part of a shared vulnerability factor associated with the underlying neurobiology of neurodevelopmental disorders such as ADHD and ASD.
Collapse
Affiliation(s)
- Djenifer B Kappel
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil.,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Jaqueline B Schuch
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil.,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Graduate Program in Biomedical Gerontology, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Diego L Rovaris
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil.,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Bruna S da Silva
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil.,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Diana Müller
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil.,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Vitor Breda
- ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Stefania P Teche
- ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rudimar S Riesgo
- Child Neurology Unit, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lavínia Schüler-Faccini
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil
| | - Luís A Rohde
- ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,National Institute of Developmental Psychiatry for Children and Adolescents, Porto Alegre, Brazil
| | - Eugenio H Grevet
- ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Claiton H D Bau
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil. .,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
82
|
Epigenetic and Neurological Impairments Associated with Early Life Exposure to Persistent Organic Pollutants. Int J Genomics 2019; 2019:2085496. [PMID: 30733955 PMCID: PMC6348822 DOI: 10.1155/2019/2085496] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/14/2018] [Accepted: 10/31/2018] [Indexed: 12/31/2022] Open
Abstract
The incidence of neurodevelopmental and neurodegenerative diseases worldwide has dramatically increased over the last decades. Although the aetiology remains uncertain, evidence is now growing that exposure to persistent organic pollutants during sensitive neurodevelopmental periods such as early life may be a strong risk factor, predisposing the individual to disease development later in life. Epidemiological studies have associated environmentally persistent organic pollutant exposure to brain disorders including neuropathies, cognitive, motor, and sensory impairments; neurodevelopmental disorders such as autism spectrum disorder (ASD) and attention-deficit hyperactivity disorder (ADHD); and neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis (ALS). In many ways, this expands the classical “Developmental Origins of Health and Disease” paradigm to include exposure to pollutants. This model has been refined over the years to give the current “three-hit” model that considers the individual's genetic factors as a first “hit.” It has an immediate interaction with the early-life exposome (including persistent organic pollutants) that can be considered to be a second “hit.” Together, these first two “hits” produce a quiescent or latent phenotype, most probably encoded in the epigenome, which has become susceptible to a third environmental “hit” in later life. It is only after the third “hit” that the increased risk of disease symptoms is crystallised. However, if the individual is exposed to a different environment in later life, they would be expected to remain healthy. In this review, we examine the effect of exposure to persistent organic pollutants and particulate matters in early life and the relationship to subsequent neurodevelopmental and neurodegenerative disorders. The roles of those environmental factors which may affect epigenetic DNA methylation and therefore influence normal neurodevelopment are then evaluated.
Collapse
|
83
|
Demontis D, Walters RK, Martin J, Mattheisen M, Als TD, Agerbo E, Baldursson G, Belliveau R, Bybjerg-Grauholm J, Bækvad-Hansen M, Cerrato F, Chambert K, Churchhouse C, Dumont A, Eriksson N, Gandal M, Goldstein JI, Grasby KL, Grove J, Gudmundsson OO, Hansen CS, Hauberg ME, Hollegaard MV, Howrigan DP, Huang H, Maller JB, Martin AR, Martin NG, Moran J, Pallesen J, Palmer DS, Pedersen CB, Pedersen MG, Poterba T, Poulsen JB, Ripke S, Robinson EB, Satterstrom FK, Stefansson H, Stevens C, Turley P, Walters GB, Won H, Wright MJ, Andreassen OA, Asherson P, Burton CL, Boomsma DI, Cormand B, Dalsgaard S, Franke B, Gelernter J, Geschwind D, Hakonarson H, Haavik J, Kranzler HR, Kuntsi J, Langley K, Lesch KP, Middeldorp C, Reif A, Rohde LA, Roussos P, Schachar R, Sklar P, Sonuga-Barke EJS, Sullivan PF, Thapar A, Tung JY, Waldman ID, Medland SE, Stefansson K, Nordentoft M, Hougaard DM, Werge T, Mors O, Mortensen PB, Daly MJ, Faraone SV, Børglum AD, Neale BM. Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder. Nat Genet 2019; 51:63-75. [PMID: 30478444 DOI: 10.1101/145581] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/28/2018] [Indexed: 05/27/2023]
Abstract
Attention deficit/hyperactivity disorder (ADHD) is a highly heritable childhood behavioral disorder affecting 5% of children and 2.5% of adults. Common genetic variants contribute substantially to ADHD susceptibility, but no variants have been robustly associated with ADHD. We report a genome-wide association meta-analysis of 20,183 individuals diagnosed with ADHD and 35,191 controls that identifies variants surpassing genome-wide significance in 12 independent loci, finding important new information about the underlying biology of ADHD. Associations are enriched in evolutionarily constrained genomic regions and loss-of-function intolerant genes and around brain-expressed regulatory marks. Analyses of three replication studies: a cohort of individuals diagnosed with ADHD, a self-reported ADHD sample and a meta-analysis of quantitative measures of ADHD symptoms in the population, support these findings while highlighting study-specific differences on genetic overlap with educational attainment. Strong concordance with GWAS of quantitative population measures of ADHD symptoms supports that clinical diagnosis of ADHD is an extreme expression of continuous heritable traits.
Collapse
Affiliation(s)
- Ditte Demontis
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Raymond K Walters
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joanna Martin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- MRC Centre for Neuropsychiatric Genetics & Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Manuel Mattheisen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Thomas D Als
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Esben Agerbo
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Gísli Baldursson
- Department of Child and Adolescent Psychiatry, National University Hospital, Reykjavik, Iceland
| | - Rich Belliveau
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonas Bybjerg-Grauholm
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Marie Bækvad-Hansen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Felecia Cerrato
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kimberly Chambert
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Claire Churchhouse
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashley Dumont
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Michael Gandal
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jacqueline I Goldstein
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jakob Grove
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Olafur O Gudmundsson
- Department of Child and Adolescent Psychiatry, National University Hospital, Reykjavik, Iceland
- deCODE genetics/Amgen, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Christine S Hansen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Institute of Biological Psychiatry, MHC Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
| | - Mads Engel Hauberg
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Mads V Hollegaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Daniel P Howrigan
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julian B Maller
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genomics plc, Oxford, UK
| | - Alicia R Martin
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jennifer Moran
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonatan Pallesen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Duncan S Palmer
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carsten Bøcker Pedersen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Marianne Giørtz Pedersen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Timothy Poterba
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesper Buchhave Poulsen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Stephan Ripke
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
| | - Elise B Robinson
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard Chan School of Public Health, Boston, MA, USA
| | - F Kyle Satterstrom
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Christine Stevens
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Patrick Turley
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - G Bragi Walters
- deCODE genetics/Amgen, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Hyejung Won
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Ole A Andreassen
- NORMENT KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Philip Asherson
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Christie L Burton
- Psychiatry, Neurosciences and Mental Health, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Dorret I Boomsma
- Department of Biological Psychology, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
- EMGO Institute for Health and Care Research, Amsterdam, The Netherlands
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues de Llobregat, Barcelona, Catalonia, Spain
| | - Søren Dalsgaard
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Barbara Franke
- Departments of Human Genetics (855) and Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Joel Gelernter
- Department of Psychiatry, Genetics, and Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Daniel Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, The Children´s Hospital of Philadelphia, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jan Haavik
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
- Haukeland University Hospital, Bergen, Norway
| | - Henry R Kranzler
- Department of Psychiatry, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Veterans Integrated Service Network (VISN4) Mental Illness Research, Education, and Clinical Center (MIRECC), Crescenz VA Medical Center, Philadephia, PA, USA
| | - Jonna Kuntsi
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Kate Langley
- MRC Centre for Neuropsychiatric Genetics & Genomics, School of Medicine, Cardiff University, Cardiff, UK
- School of Psychology, Cardiff University, Cardiff, UK
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
- Department of Neuroscience, School for Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, The Netherlands
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Christel Middeldorp
- Department of Biological Psychology, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
- Child Health Research Centre, University of Queensland, Brisbane, Australia
- Child and Youth Mental Health Service, Children's Health Queensland Hospital and Health Service, Brisbane, Australia
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Luis Augusto Rohde
- Department of Psychiatry, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- ADHD Outpatient Clinic, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, New York, USA
| | - Russell Schachar
- Psychiatry, Neurosciences and Mental Health, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Pamela Sklar
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Anita Thapar
- MRC Centre for Neuropsychiatric Genetics & Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | | | - Irwin D Waldman
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kari Stefansson
- deCODE genetics/Amgen, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Merete Nordentoft
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Mental Health Services in the Capital Region of Denmark, Mental Health Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - David M Hougaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Thomas Werge
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Institute of Biological Psychiatry, MHC Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ole Mors
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Preben Bo Mortensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Stephen V Faraone
- Departments of Psychiatry and Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Anders D Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark.
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark.
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
84
|
Demontis D, Walters RK, Martin J, Mattheisen M, Als TD, Agerbo E, Baldursson G, Belliveau R, Bybjerg-Grauholm J, Bækvad-Hansen M, Cerrato F, Chambert K, Churchhouse C, Dumont A, Eriksson N, Gandal M, Goldstein JI, Grasby KL, Grove J, Gudmundsson OO, Hansen CS, Hauberg ME, Hollegaard MV, Howrigan DP, Huang H, Maller JB, Martin AR, Martin NG, Moran J, Pallesen J, Palmer DS, Pedersen CB, Pedersen MG, Poterba T, Poulsen JB, Ripke S, Robinson EB, Satterstrom FK, Stefansson H, Stevens C, Turley P, Walters GB, Won H, Wright MJ, Andreassen OA, Asherson P, Burton CL, Boomsma DI, Cormand B, Dalsgaard S, Franke B, Gelernter J, Geschwind D, Hakonarson H, Haavik J, Kranzler HR, Kuntsi J, Langley K, Lesch KP, Middeldorp C, Reif A, Rohde LA, Roussos P, Schachar R, Sklar P, Sonuga-Barke EJS, Sullivan PF, Thapar A, Tung JY, Waldman ID, Medland SE, Stefansson K, Nordentoft M, Hougaard DM, Werge T, Mors O, Mortensen PB, Daly MJ, Faraone SV, Børglum AD, Neale BM. Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder. Nat Genet 2019; 51:63-75. [PMID: 30478444 PMCID: PMC6481311 DOI: 10.1038/s41588-018-0269-7] [Citation(s) in RCA: 1229] [Impact Index Per Article: 245.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/28/2018] [Indexed: 02/07/2023]
Abstract
Attention deficit/hyperactivity disorder (ADHD) is a highly heritable childhood behavioral disorder affecting 5% of children and 2.5% of adults. Common genetic variants contribute substantially to ADHD susceptibility, but no variants have been robustly associated with ADHD. We report a genome-wide association meta-analysis of 20,183 individuals diagnosed with ADHD and 35,191 controls that identifies variants surpassing genome-wide significance in 12 independent loci, finding important new information about the underlying biology of ADHD. Associations are enriched in evolutionarily constrained genomic regions and loss-of-function intolerant genes and around brain-expressed regulatory marks. Analyses of three replication studies: a cohort of individuals diagnosed with ADHD, a self-reported ADHD sample and a meta-analysis of quantitative measures of ADHD symptoms in the population, support these findings while highlighting study-specific differences on genetic overlap with educational attainment. Strong concordance with GWAS of quantitative population measures of ADHD symptoms supports that clinical diagnosis of ADHD is an extreme expression of continuous heritable traits.
Collapse
Affiliation(s)
- Ditte Demontis
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Raymond K Walters
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joanna Martin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- MRC Centre for Neuropsychiatric Genetics & Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Manuel Mattheisen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Thomas D Als
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Esben Agerbo
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Gísli Baldursson
- Department of Child and Adolescent Psychiatry, National University Hospital, Reykjavik, Iceland
| | - Rich Belliveau
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonas Bybjerg-Grauholm
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Marie Bækvad-Hansen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Felecia Cerrato
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kimberly Chambert
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Claire Churchhouse
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashley Dumont
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Michael Gandal
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jacqueline I Goldstein
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jakob Grove
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Olafur O Gudmundsson
- Department of Child and Adolescent Psychiatry, National University Hospital, Reykjavik, Iceland
- deCODE genetics/Amgen, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Christine S Hansen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Institute of Biological Psychiatry, MHC Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
| | - Mads Engel Hauberg
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Mads V Hollegaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Daniel P Howrigan
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julian B Maller
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genomics plc, Oxford, UK
| | - Alicia R Martin
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jennifer Moran
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonatan Pallesen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Duncan S Palmer
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carsten Bøcker Pedersen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Marianne Giørtz Pedersen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Timothy Poterba
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesper Buchhave Poulsen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Stephan Ripke
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
| | - Elise B Robinson
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard Chan School of Public Health, Boston, MA, USA
| | - F Kyle Satterstrom
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Christine Stevens
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Patrick Turley
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - G Bragi Walters
- deCODE genetics/Amgen, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Hyejung Won
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | | | | | | | - Ole A Andreassen
- NORMENT KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Philip Asherson
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Christie L Burton
- Psychiatry, Neurosciences and Mental Health, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Dorret I Boomsma
- Department of Biological Psychology, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
- EMGO Institute for Health and Care Research, Amsterdam, The Netherlands
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues de Llobregat, Barcelona, Catalonia, Spain
| | - Søren Dalsgaard
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Barbara Franke
- Departments of Human Genetics (855) and Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Joel Gelernter
- Department of Psychiatry, Genetics, and Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Daniel Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, The Children´s Hospital of Philadelphia, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jan Haavik
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
- Haukeland University Hospital, Bergen, Norway
| | - Henry R Kranzler
- Department of Psychiatry, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Veterans Integrated Service Network (VISN4) Mental Illness Research, Education, and Clinical Center (MIRECC), Crescenz VA Medical Center, Philadephia, PA, USA
| | - Jonna Kuntsi
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Kate Langley
- MRC Centre for Neuropsychiatric Genetics & Genomics, School of Medicine, Cardiff University, Cardiff, UK
- School of Psychology, Cardiff University, Cardiff, UK
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
- Department of Neuroscience, School for Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, The Netherlands
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Christel Middeldorp
- Department of Biological Psychology, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
- Child Health Research Centre, University of Queensland, Brisbane, Australia
- Child and Youth Mental Health Service, Children's Health Queensland Hospital and Health Service, Brisbane, Australia
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Luis Augusto Rohde
- Department of Psychiatry, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- ADHD Outpatient Clinic, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, New York, USA
| | - Russell Schachar
- Psychiatry, Neurosciences and Mental Health, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Pamela Sklar
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Anita Thapar
- MRC Centre for Neuropsychiatric Genetics & Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | | | - Irwin D Waldman
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kari Stefansson
- deCODE genetics/Amgen, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Merete Nordentoft
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Mental Health Services in the Capital Region of Denmark, Mental Health Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - David M Hougaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Thomas Werge
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Institute of Biological Psychiatry, MHC Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ole Mors
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Preben Bo Mortensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Stephen V Faraone
- Departments of Psychiatry and Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Anders D Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark.
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark.
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
85
|
Mehta D, Czamara D. GWAS of Behavioral Traits. Curr Top Behav Neurosci 2019; 42:1-34. [PMID: 31407241 DOI: 10.1007/7854_2019_105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Over the past decade, genome-wide association studies (GWAS) have evolved into a powerful tool to investigate genetic risk factors for human diseases via a hypothesis-free scan of the genome. The success of GWAS for psychiatric disorders and behavioral traits have been somewhat mixed, partly owing to the complexity and heterogeneity of these traits. Significant progress has been made in the last few years in the development and implementation of complex statistical methods and algorithms incorporating GWAS. Such advanced statistical methods applied to GWAS hits in combination with incorporation of different layers of genomics data have catapulted the search for novel genes for behavioral traits and improved our understanding of the complex polygenic architecture of these traits.This chapter will give a brief overview on GWAS and statistical methods currently used in GWAS. The chapter will focus on reviewing the current literature and highlight some of the most important GWAS on psychiatric and other behavioral traits and will conclude with a discussion on future directions.
Collapse
Affiliation(s)
- Divya Mehta
- School of Psychology and Counselling, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.
| | - Darina Czamara
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
86
|
Hoffman GE, Schrode N, Flaherty E, Brennand KJ. New considerations for hiPSC-based models of neuropsychiatric disorders. Mol Psychiatry 2019; 24:49-66. [PMID: 29483625 PMCID: PMC6109625 DOI: 10.1038/s41380-018-0029-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/17/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023]
Abstract
The development of human-induced pluripotent stem cells (hiPSCs) has made possible patient-specific modeling across the spectrum of human disease. Here, we discuss recent advances in psychiatric genomics and post-mortem studies that provide critical insights concerning cell-type composition and sample size that should be considered when designing hiPSC-based studies of complex genetic disease. We review recent hiPSC-based models of SZ, in light of our new understanding of critical power limitations in the design of hiPSC-based studies of complex genetic disorders. Three possible solutions are a movement towards genetically stratified cohorts of rare variant patients, application of CRISPR technologies to engineer isogenic neural cells to study the impact of common variants, and integration of advanced genetics and hiPSC-based datasets in future studies. Overall, we emphasize that to advance the reproducibility and relevance of hiPSC-based studies, stem cell biologists must contemplate statistical and biological considerations that are already well accepted in the field of genetics. We conclude with a discussion of the hypothesis of biological convergence of disease-through molecular, cellular, circuit, and patient level phenotypes-and how this might emerge through hiPSC-based studies.
Collapse
Affiliation(s)
- Gabriel E Hoffman
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nadine Schrode
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Erin Flaherty
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kristen J Brennand
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
87
|
Hawi Z, Yates H, Pinar A, Arnatkeviciute A, Johnson B, Tong J, Pugsley K, Dark C, Pauper M, Klein M, Heussler HS, Hiscock H, Fornito A, Tiego J, Finlay A, Vance A, Gill M, Kent L, Bellgrove MA. A case-control genome-wide association study of ADHD discovers a novel association with the tenascin R (TNR) gene. Transl Psychiatry 2018; 8:284. [PMID: 30563984 PMCID: PMC6298965 DOI: 10.1038/s41398-018-0329-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/08/2018] [Indexed: 11/29/2022] Open
Abstract
It is well-established that there is a strong genetic contribution to the aetiology of attention deficit hyperactivity disorder (ADHD). Here, we employed a hypothesis-free genome-wide association study (GWAS) design in a sample of 480 clinical childhood ADHD cases and 1208 controls to search for novel genetic risk loci for ADHD. DNA was genotyped using Illumina's Human Infinium PsychArray-24v1.2., and the data were subsequently imputed to the 1000 Genomes reference panel. Rigorous quality control and pruning of genotypes at both individual subject and single nucleotide polymorphism (SNP) levels was performed. Polygenic risk score (PGRS) analysis revealed that ADHD case-control status was explained by genetic risk for ADHD, but no other major psychiatric disorders. Logistic regression analysis was performed genome-wide to test the association between SNPs and ADHD case-control status. We observed a genome-wide significant association (p = 3.15E-08) between ADHD and rs6686722, mapped to the Tenascin R (TNR) gene. Members of this gene family are extracellular matrix glycoproteins that play a role in neural cell adhesion and neurite outgrowth. Suggestive evidence of associations with ADHD was observed for an additional 111 SNPs (⩽9.91E-05). Although intriguing, the association between DNA variation in the TNR gene and ADHD should be viewed as preliminary given the small sample size of this discovery dataset.
Collapse
Affiliation(s)
- Ziarih Hawi
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia.
| | - Hannah Yates
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| | - Ari Pinar
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| | - Aurina Arnatkeviciute
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| | - Beth Johnson
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| | - Janette Tong
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| | - Kealan Pugsley
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| | - Callum Dark
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| | - Marc Pauper
- Departments of Human Genetics, and Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marieke Klein
- Departments of Human Genetics, and Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Helen S Heussler
- Mater Research Institute, University of Queensland and Children's Health Queensland, South Brisbane, Australia
| | - Harriet Hiscock
- Pediatrics Royal Children's Hospital, Murdoch Children's Institute, Melbourne, Australia
| | - Alex Fornito
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| | - Jeggan Tiego
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| | - Amy Finlay
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| | - Alasdair Vance
- The Royal Children's Hospital, University of Melbourne, Victoria, Australia
| | - Michael Gill
- Department of Psychiatry, Trinity College, Dublin, Ireland
| | - Lindsey Kent
- School of Medicine, University of St Andrews, St. Andrews, Scotland, UK
| | - Mark A Bellgrove
- School of Psychological Sciences and Monash Institute for Cognitive and Clinical Neurosciences (MICCN), Monash University, Melbourne, Australia
| |
Collapse
|
88
|
Cortese S, Coghill D. Twenty years of research on attention-deficit/hyperactivity disorder (ADHD): looking back, looking forward. EVIDENCE-BASED MENTAL HEALTH 2018; 21:173-176. [PMID: 30301823 PMCID: PMC10270437 DOI: 10.1136/ebmental-2018-300050] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/06/2018] [Accepted: 09/13/2018] [Indexed: 01/07/2023]
Abstract
In this clinical review we summarise what in our view have been some the most important advances in the past two decades, in terms of diagnostic definition, epidemiology, genetics and environmental causes, neuroimaging/cognition and treatment of attention-deficit/hyperactivity disorder (ADHD), including: (1) the most recent changes to the diagnostic criteria in the Diagnostic and Statistical Manual of Mental Disorders and International Classification of Diseases; (2) meta-analytic evidence showing that, after accounting for diagnostic methods, the rates of ADHD are fairly consistent across Western countries; (3) the recent finding of the first genome-wide significant risk loci for ADHD; (4) the paradigm shift in the pathophysiological conceptualisation of ADHD from alterations in individual brain regions to a complex dysfunction in brain networks; (5) evidence supporting the short-term efficacy of ADHD pharmacological treatments, with a different profile of efficacy and tolerability in children/adolescents versus adults; (6) a series of meta-analyses showing that, while non-pharmacological treatment may not be effective to target ADHD core symptoms, some of them effectively address ADHD-related impairments (such as oppositional behaviours for parent training and working memory deficits for cognitive training). We also discuss key priorities for future research in each of these areas of investigation. Overall, while many research questions have been answered, many others need to be addressed. Strengthening multidisciplinary collaborations, relying on large data sets in the spirit of Open Science and supporting research in less advantaged countries will be key to face the challenges ahead.
Collapse
Affiliation(s)
- Samuele Cortese
- Academic Unit of Psychology, Center for Innovation in Mental Health, University of Southampton, Southampton, UK
- Clinical and Experimental Sciences (CNS and Psychiatry), Faculty of Medicine, University of Southampton, Southampton, UK
- Solent NHS Trust, Southampton, UK
- New York University Child Study Center, New York City, New York, USA
- Division of Psychiatry and Applied Psychology, School of Medicine, University of Nottingham, Nottingham, UK
| | - David Coghill
- Departments of Paediatrics and Psychiatry, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
- Royal Children’s Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
89
|
Schäfer N, Friedrich M, Jørgensen ME, Kollert S, Koepsell H, Wischmeyer E, Lesch KP, Geiger D, Döring F. Functional analysis of a triplet deletion in the gene encoding the sodium glucose transporter 3, a potential risk factor for ADHD. PLoS One 2018; 13:e0205109. [PMID: 30286162 PMCID: PMC6171906 DOI: 10.1371/journal.pone.0205109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 09/19/2018] [Indexed: 12/19/2022] Open
Abstract
Sodium-glucose transporters (SGLT) belong to the solute carrier 5 family, which is characterized by sodium dependent transport of sugars and other solutes. In contrast, the human SGLT3 (hSGLT3) isoform, encoded by SLC5A4, acts as a glucose sensor that does not transport sugar but induces membrane depolarization by Na+ currents upon ligand binding. Whole-exome sequencing (WES) of several extended pedigrees with high density of attention-deficit/hyperactivity disorder (ADHD) identified a triplet ATG deletion in SLC5A4 leading to a single amino acid loss (ΔM500) in the hSGLT3 protein imperfectly co-segregating with the clinical phenotype of ADHD. Since mutations in homologous domains of hSGLT1 and hSGLT2 were found to affect intestinal and renal function, respectively, we analyzed the functional properties of hSGLT3[wt] and [ΔM500] by voltage clamp and current clamp recordings from cRNA-injected Xenopus laevis oocytes. The cation conductance of hSGLT3[wt] was activated by application of glucose or the specific agonist 1-desoxynojirimycin (DNJ) as revealed by inward currents in the voltage clamp configuration and cell depolarization in the current clamp mode. Almost no currents and changes in membrane potential were observed when glucose or DNJ were applied to hSGLT3[ΔM500]-injected oocytes, demonstrating a loss of function by this amino acid deletion in hSGLT3. To monitor membrane targeting of wt and mutant hSGLT3, fusion constructs with YFP were generated, heterologously expressed in Xenopus laevis oocytes and analyzed for membrane fluorescence by confocal microscopy. In comparison to hSGLT3[wt] the fluorescent signal of mutant [ΔM500] was reduced by 43% indicating that the mutant phenotype might mainly result from inaccurate membrane targeting. As revealed by homology modeling, residue M500 is located in TM11 suggesting that in addition to the core structure (TM1-TM10) of the transporter, the surrounding TMs are equally crucial for transport/sensor function. In conclusion, our findings indicate that the deletion [ΔM500] in hSGLT3 inhibits membrane targeting and thus largely disrupts glucose-induced sodium conductance, which may, in interaction with other ADHD risk-related gene variants, influence the risk for ADHD in deletion carriers.
Collapse
Affiliation(s)
- Nadine Schäfer
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| | - Maximilian Friedrich
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Morten Egevang Jørgensen
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| | - Sina Kollert
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
- Division of Molecular Electrophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Hermann Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| | - Erhard Wischmeyer
- Division of Molecular Electrophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health,University Hospital of Würzburg, Würzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Neuroscience, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Dietmar Geiger
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| | - Frank Döring
- Division of Molecular Electrophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health,University Hospital of Würzburg, Würzburg, Germany
- * E-mail:
| |
Collapse
|
90
|
Franke B, Michelini G, Asherson P, Banaschewski T, Bilbow A, Buitelaar JK, Cormand B, Faraone SV, Ginsberg Y, Haavik J, Kuntsi J, Larsson H, Lesch KP, Ramos-Quiroga JA, Réthelyi JM, Ribases M, Reif A. Live fast, die young? A review on the developmental trajectories of ADHD across the lifespan. Eur Neuropsychopharmacol 2018; 28:1059-1088. [PMID: 30195575 PMCID: PMC6379245 DOI: 10.1016/j.euroneuro.2018.08.001] [Citation(s) in RCA: 321] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 06/25/2018] [Accepted: 08/07/2018] [Indexed: 02/07/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is highly heritable and the most common neurodevelopmental disorder in childhood. In recent decades, it has been appreciated that in a substantial number of cases the disorder does not remit in puberty, but persists into adulthood. Both in childhood and adulthood, ADHD is characterised by substantial comorbidity including substance use, depression, anxiety, and accidents. However, course and symptoms of the disorder and the comorbidities may fluctuate and change over time, and even age of onset in childhood has recently been questioned. Available evidence to date is poor and largely inconsistent with regard to the predictors of persistence versus remittance. Likewise, the development of comorbid disorders cannot be foreseen early on, hampering preventive measures. These facts call for a lifespan perspective on ADHD from childhood to old age. In this selective review, we summarise current knowledge of the long-term course of ADHD, with an emphasis on clinical symptom and cognitive trajectories, treatment effects over the lifespan, and the development of comorbidities. Also, we summarise current knowledge and important unresolved issues on biological factors underlying different ADHD trajectories. We conclude that a severe lack of knowledge on lifespan aspects in ADHD still exists for nearly every aspect reviewed. We encourage large-scale research efforts to overcome those knowledge gaps through appropriately granular longitudinal studies.
Collapse
Affiliation(s)
- Barbara Franke
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Giorgia Michelini
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Social, Genetic & Developmental Psychiatry Centre, London, UK
| | - Philip Asherson
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Social, Genetic & Developmental Psychiatry Centre, London, UK
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Andrea Bilbow
- Attention Deficit Disorder Information and Support Service (ADDISS), Edgware, UK; ADHD-Europe, Brussels, Belgium
| | - Jan K Buitelaar
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Nijmegen, The Netherlands
| | - Bru Cormand
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, State University of New York Upstate Medical University, New York, USA; K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ylva Ginsberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Jan Haavik
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway; Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Jonna Kuntsi
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Social, Genetic & Developmental Psychiatry Centre, London, UK
| | - Henrik Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Department of Translational Neuroscience, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - J Antoni Ramos-Quiroga
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Catalonia, Spain; Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Barcelona, Catalonia, Spain; Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - János M Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; MTA-SE NAP-B Molecular Psychiatry Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Marta Ribases
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Catalonia, Spain; Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Barcelona, Catalonia, Spain
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
91
|
Elwasify M, Elwasify M, Mostafa M, Saleh E, Hamdey I, Barakat D. Methylenetetrahydrofolate reductase gene polymorphisms and executive functions in a sample of Egyptian children with attention-deficit hyperactivity disorder. MIDDLE EAST CURRENT PSYCHIATRY 2018. [DOI: 10.1097/01.xme.0000542437.67434.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
92
|
Grimm O, Kittel-Schneider S, Reif A. Recent developments in the genetics of attention-deficit hyperactivity disorder. Psychiatry Clin Neurosci 2018; 72:654-672. [PMID: 29722101 DOI: 10.1111/pcn.12673] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/01/2018] [Indexed: 12/19/2022]
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a developmental psychiatric disorder that affects children and adults. ADHD is one of the psychiatric disorders with the strongest genetic basis according to familial, twin, and single nucleotide polymorphisms (SNP)-based epidemiological studies. In this review, we provide an update of recent insights into the genetic basis of ADHD. We discuss recent progress from genome-wide association studies (GWAS) looking at common variants as well as rare copy number variations. New analysis of gene groups, so-called functional ontologies, provide some insight into the gene networks afflicted, pointing to the role of neurodevelopmentally expressed gene networks. Bioinformatic methods, such as functional enrichment analysis and protein-protein network analysis, are used to highlight biological processes of likely relevance to the etiology of ADHD. Additionally, copy number variations seem to map on important pathways implicated in synaptic signaling and neurodevelopment. While some candidate gene associations of, for example, neurotransmitter receptors and signaling, have been replicated, they do not seem to explain significant variance in recent GWAS. We discuss insights from recent case-control SNP-GWAS that have presented the first whole-genome significant SNP in ADHD.
Collapse
Affiliation(s)
- Oliver Grimm
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
93
|
Zheng J, Chen YH. [Research advances in pathogenesis of attention deficit hyperactivity disorder]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:775-780. [PMID: 30210033 PMCID: PMC7389180 DOI: 10.7499/j.issn.1008-8830.2018.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/07/2018] [Indexed: 06/08/2023]
Abstract
Both of genetic and environmental factors play important roles in the pathogenesis of attention deficit hyperactivity disorder (ADHD), and genetic factors can increase the susceptibility of individuals to environmental risk factors. There are extensive and various structural and functional abnormalities of the brain in patients with ADHD. Given the close functional relationship between brain areas, exploration has also been expanded to the dysfunction of brain network in recent years. As for the biochemical mechanism underlying ADHD, monoamine neurotransmitters are still most valued, and abnormalities of brain-derived neurotrophic factors and glutamic acid/γ-aminobutyric acid imbalance may also be present. Due to the abnormal neuroendocrine function and connectivity between brain areas caused by the synergistic effect of genetic and environmental factors, the prefrontal cortex loses control of the lower brain areas, so that the basal ganglia and amygdala affect normal behavioral and emotional reactions. Dysfunction of the endocrine axes may further aggravate neuroendocrine disorder. The above process may eventually lead to changes in brain structure and function, which may be associated with the development of ADHD. However, considering the heterogeneity of ADHD, its pathological process may not be the same, and the exact mechanism needs to be further clarified.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Pediatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| | | |
Collapse
|
94
|
Pharmacogenetics predictors of methylphenidate efficacy in childhood ADHD. Mol Psychiatry 2018; 23:1929-1936. [PMID: 29230023 PMCID: PMC7039663 DOI: 10.1038/mp.2017.234] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/22/2017] [Accepted: 10/10/2017] [Indexed: 11/08/2022]
Abstract
Stimulant medication has long been effective in treating attention-deficit/hyperactivity disorder (ADHD) and is currently the first-line pharmacological treatment for children. Both methylphenidate and amphetamine modulate extracellular catecholamine levels through interaction with dopaminergic, adrenergic and serotonergic system components; it is therefore likely that catecholaminergic molecular components influence the effects of ADHD treatment. Using meta-analysis, we sought to identify predictors of pharmacotherapy to further the clinical implementation of personalized medicine. We identified 36 studies (3647 children) linking the effectiveness of methylphenidate treatment with DNA variants. Pooled-data revealed a statistically significant association between single nucleotide polymorphisms (SNPs) rs1800544 ADRA2A (odds ratio: 1.69; confidence interval: 1.12-2.55), rs4680 COMT (odds ratio (OR): 1.40; confidence interval: 1.04-1.87), rs5569 SLC6A2 (odds ratio: 1.73; confidence interval: 1.26-2.37) and rs28386840 SLC6A2 (odds ratio: 2.93; confidence interval: 1.76-4.90), and, repeat variants variable number tandem repeat (VNTR) 4 DRD4 (odds ratio: 1.66; confidence interval: 1.16-2.37) and VNTR 10 SLC6A3 (odds ratio: 0.74; confidence interval: 0.60-0.90), whereas the following variants were not statistically significant: rs1947274 LPHN3 (odds ratio: 0.95; confidence interval: 0.71-1.26), rs5661665 LPHN3 (odds ratio: 1.07; confidence interval: 0.84-1.37) and VNTR 7 DRD4 (odds ratio: 0.68; confidence interval: 0.47-1.00). Funnel plot asymmetry among SLC6A3 studies was identified and attributed largely to small study effects. Egger's regression test and Duval and Tweedie's 'trim and fill' were used to examine and correct for publication bias. These findings have major implications for advancing our therapeutic approach to childhood ADHD treatment.
Collapse
|
95
|
Hong JH, Hwang IW, Lim MH, Kwon HJ, Jin HJ. Genetic associations between ADHD and dopaminergic genes (DAT1 and DRD4) VNTRs in Korean children. Genes Genomics 2018; 40:1309-1317. [PMID: 30099719 DOI: 10.1007/s13258-018-0726-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/08/2018] [Indexed: 01/01/2023]
Abstract
It is well known that dopaminergic genes affect the development of attention deficit hyperactivity disorder (ADHD) in various populations. Many studies have shown that variable number tandem repeats (VNTRs) located within the 3'-untranslated region of DAT1 and in exon 3 of DRD4 are associated with ADHD development; however, these results were inconsistent. Therefore, we investigated the genetic association between two VNTRs and ADHD in Korean children. We determined the VNTRs using PCR. We examined genotype and allele frequency differences between the experimental and control groups, along with the odds ratios, using Chi square and exact tests. We observed a significant association between the children with ADHD and the control group in the 10R/10R genotype of DAT1 VNTRs (p = 0.025). In addition, the 11R allele of DAT1 VNTRs showed a higher frequency in the control group than in the ADHD group (p = 0.023). Also, the short repeat (without 11R) and long repeat alleles (including 11R) were associated with ADHD (p < 0.05). The analysis of DRD4 VNTRs revealed that the 2R allele is associated with ADHD (p = 0.025). A significant result was also observed in long and short repeats (p < 0.05). Additionally, ADHD subtypes showed that the DRD4 VNTRs are associated with combined and hyperactive-impulsive subtype groups (p < 0.05). Therefore, our results suggest that DAT1 VNTRs and DRD4 VNTRs play a role in the genetic etiology of ADHD in Korean children.
Collapse
Affiliation(s)
- Jun Ho Hong
- Department of Biological Sciences, College of Natural Science, Dankook University, Cheonan, 330-714, Republic of Korea
| | - In Wook Hwang
- Department of Biological Sciences, College of Natural Science, Dankook University, Cheonan, 330-714, Republic of Korea
- Enviromental Health Center, Dankook Medical Hospital, Cheonan, Republic of Korea
| | - Myung Ho Lim
- Department of Psychology and Psychotherapy, College of Health Sciences, Dankook University, Cheonan, Republic of Korea
- Enviromental Health Center, Dankook Medical Hospital, Cheonan, Republic of Korea
| | - Ho Jang Kwon
- Department of Preventive Medicine, College of Medicine, Dankook University, Cheonan, Republic of Korea
- Enviromental Health Center, Dankook Medical Hospital, Cheonan, Republic of Korea
| | - Han Jun Jin
- Department of Biological Sciences, College of Natural Science, Dankook University, Cheonan, 330-714, Republic of Korea.
- Enviromental Health Center, Dankook Medical Hospital, Cheonan, Republic of Korea.
| |
Collapse
|
96
|
Prehn-Kristensen A, Zimmermann A, Tittmann L, Lieb W, Schreiber S, Baving L, Fischer A. Reduced microbiome alpha diversity in young patients with ADHD. PLoS One 2018; 13:e0200728. [PMID: 30001426 PMCID: PMC6042771 DOI: 10.1371/journal.pone.0200728] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 07/02/2018] [Indexed: 12/16/2022] Open
Abstract
ADHD is a psychiatric disorder which is characterized by hyperactivity, impulsivity and attention problems. Due to recent findings of microbial involvement in other psychiatric disorders like autism and depression, a role of the gut microbiota in ADHD pathogenesis is assumed but has not yet been investigated. In this study, the gut microbiota of 14 male ADHD patients (mean age: 11.9 yrs.) and 17 male controls (mean age: 13.1 yrs.) was examined via next generation sequencing of 16S rDNA and analyzed for diversity and biomarkers. We found that the microbial diversity (alpha diversity) was significantly decreased in ADHD patients compared to controls (pShannon = 0.036) and that the composition (beta diversity) differed significantly between patients and controls (pANOSIM = 0.033, pADONIS = 0.006, pbetadisper = 0.002). In detail, the bacterial family Prevotellacae was associated with controls, while patients with ADHD showed elevated levels of Bacteroidaceae, and both Neisseriaceae and Neisseria spec. were found as possible biomarkers for juvenile ADHD. Our results point to a possible link of certain microbiota with ADHD, with Neisseria spec. being a very promising ADHD-associated candidate. This finding provides the basis for a systematic, longitudinal assessment of the role of the gut microbiome in ADHD, yielding promising potential for both prevention and therapeutic intervention.
Collapse
Affiliation(s)
- Alexander Prehn-Kristensen
- Department of Child and Adolescent Psychiatry and Psychotherapy, Centre for Integrative Psychiatry, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Alexandra Zimmermann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Centre for Integrative Psychiatry, University Hospital Schleswig-Holstein, Kiel, Germany
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lukas Tittmann
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Wolfgang Lieb
- Institute for Epidemiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Kiel, Germany
- Clinic of Internal Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lioba Baving
- Department of Child and Adolescent Psychiatry and Psychotherapy, Centre for Integrative Psychiatry, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Annegret Fischer
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
97
|
Gao Q, Liu L, Li HM, Tang YL, Chen Y, Wang YF, Qian QJ. Interaction Between Season of Birth and COMT Val158Met (rs4680) in ADHD in a Large Sample of Chinese Han Participants. J Atten Disord 2018; 22:886-895. [PMID: 26486601 DOI: 10.1177/1087054715608441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To investigate the interaction between catechol-O-methyltransferase gene (COMT) Val108/158Met (rs4680) and season of birth (SOB) on ADHD and its symptoms. METHOD We conducted transmission disequilibrium tests (TDTs) in 976 trios, then further performed the above analyses in subgroups according to SOB. Quantitative analyses were performed for ADHD symptoms evaluated by ADHD Rating Scale-IV in 1,530 ADHD cases. RESULTS Overall, there was no association between COMT and ADHD. After stratification, we found an increased transmission of the Val allele in the trios born in spring, while a decreased transmission was observed in the autumn months. We also observed a significant interaction between Val108/158Met and SOB on ADHD symptoms. Among those born in spring, Met carriers had milder ADHD symptoms compared with Val homozygotes, whereas opposite association was found in those born in autumn. CONCLUSION Our study provided evidence for the modifying effect of SOB on the association between COMT and ADHD along with its symptoms.
Collapse
Affiliation(s)
- Qian Gao
- 1 Peking University Sixth Hospital, Beijing, China.,2 Ministry of Health, Beijing, China
| | - Lu Liu
- 1 Peking University Sixth Hospital, Beijing, China.,2 Ministry of Health, Beijing, China
| | - Hai-Mei Li
- 1 Peking University Sixth Hospital, Beijing, China.,2 Ministry of Health, Beijing, China
| | - Yi-Lang Tang
- 3 Emory University School of Medicine, Atlanta, Georgia
| | - Yun Chen
- 1 Peking University Sixth Hospital, Beijing, China.,2 Ministry of Health, Beijing, China
| | - Yu-Feng Wang
- 1 Peking University Sixth Hospital, Beijing, China.,2 Ministry of Health, Beijing, China
| | - Qiu-Jin Qian
- 1 Peking University Sixth Hospital, Beijing, China.,2 Ministry of Health, Beijing, China
| |
Collapse
|
98
|
Moretto E, Murru L, Martano G, Sassone J, Passafaro M. Glutamatergic synapses in neurodevelopmental disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:328-342. [PMID: 28935587 DOI: 10.1016/j.pnpbp.2017.09.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/28/2017] [Accepted: 09/16/2017] [Indexed: 12/22/2022]
Abstract
Neurodevelopmental disorders (NDDs) are a group of diseases whose symptoms arise during childhood or adolescence and that impact several higher cognitive functions such as learning, sociability and mood. Accruing evidence suggests that a shared pathogenic mechanism underlying these diseases is the dysfunction of glutamatergic synapses. We summarize present knowledge on autism spectrum disorders (ASD), intellectual disability (ID), Down syndrome (DS), Rett syndrome (RS) and attention-deficit hyperactivity disorder (ADHD), highlighting the involvement of glutamatergic synapses and receptors in these disorders. The most commonly shared defects involve α-amino-3-hydroxy-5-methyl- 4-isoxazole propionic acid receptors (AMPARs), N-methyl-d-aspartate receptors (NMDARs) and metabotropic glutamate receptors (mGluRs), whose functions are strongly linked to synaptic plasticity, affecting both cell-autonomous features as well as circuit formation. Moreover, the major scaffolding proteins and, thus, the general structure of the synapse are often deregulated in neurodevelopmental disorders, which is not surprising considering their crucial role in the regulation of glutamate receptor positioning and functioning. This convergence of defects supports the definition of neurodevelopmental disorders as a continuum of pathological manifestations, suggesting that glutamatergic synapses could be a therapeutic target to ameliorate patient symptomatology.
Collapse
Affiliation(s)
- Edoardo Moretto
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Luca Murru
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Giuseppe Martano
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Jenny Sassone
- San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy
| | - Maria Passafaro
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy.
| |
Collapse
|
99
|
Dark C, Homman-Ludiye J, Bryson-Richardson RJ. The role of ADHD associated genes in neurodevelopment. Dev Biol 2018; 438:69-83. [DOI: 10.1016/j.ydbio.2018.03.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 03/04/2018] [Accepted: 03/27/2018] [Indexed: 12/19/2022]
|
100
|
Kumar VJ, Grissom NM, McKee SE, Schoch H, Bowman N, Havekes R, Kumar M, Pickup S, Poptani H, Reyes TM, Hawrylycz M, Abel T, Nickl-Jockschat T. Linking spatial gene expression patterns to sex-specific brain structural changes on a mouse model of 16p11.2 hemideletion. Transl Psychiatry 2018; 8:109. [PMID: 29844452 PMCID: PMC5974415 DOI: 10.1038/s41398-018-0157-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 04/02/2018] [Accepted: 04/10/2018] [Indexed: 02/02/2023] Open
Abstract
Neurodevelopmental disorders, such as ASD and ADHD, affect males about three to four times more often than females. 16p11.2 hemideletion is a copy number variation that is highly associated with neurodevelopmental disorders. Previous work from our lab has shown that a mouse model of 16p11.2 hemideletion (del/+) exhibits male-specific behavioral phenotypes. We, therefore, aimed to investigate with magnetic resonance imaging (MRI), whether del/+ animals also exhibited a sex-specific neuroanatomical endophenotype. Using the Allen Mouse Brain Atlas, we analyzed the expression patterns of the 27 genes within the 16p11.2 region to identify which gene expression patterns spatially overlapped with brain structural changes. MRI was performed ex vivo and the resulting images were analyzed using Voxel-based morphometry for T1-weighted sequences and tract-based spatial statistics for diffusion-weighted images. In a subsequent step, all available in situ hybridization (ISH) maps of the genes involved in the 16p11.2 hemideletion were aligned to Waxholm space and clusters obtained by sex-specific group comparisons were analyzed to determine which gene(s) showed the highest expression in these regions. We found pronounced sex-specific changes in male animals with increased fractional anisotropy in medial fiber tracts, especially in those proximate to the striatum. Moreover, we were able to identify gene expression patterns spatially overlapping with male-specific structural changes that were associated with neurite outgrowth and the MAPK pathway. Of note, previous molecular studies have found convergent changes that point to a sex-specific dysregulation of MAPK signaling. This convergent evidence supports the idea that ISH maps can be used to meaningfully analyze imaging data sets.
Collapse
Affiliation(s)
- Vinod Jangir Kumar
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Juelich-Aachen Research Alliance Brain, Juelich/Aachen, Germany
- Max Planck Institute for Biological Cybernetics, Tubingen, Germany
| | - Nicola M Grissom
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Sarah E McKee
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Hannah Schoch
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicole Bowman
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Robbert Havekes
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Manoj Kumar
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen Pickup
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Harish Poptani
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Teresa M Reyes
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry and Behavioral Neurosciences, University of Cincinnati, Cincinnati, OH, USA
| | | | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa, IA, USA
| | - Thomas Nickl-Jockschat
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany.
- Juelich-Aachen Research Alliance Brain, Juelich/Aachen, Germany.
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa, IA, USA.
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA, USA.
| |
Collapse
|