101
|
Selvanayagam T, Walker S, Gazzellone MJ, Kellam B, Cytrynbaum C, Stavropoulos DJ, Li P, Birken CS, Hamilton J, Weksberg R, Scherer SW. Genome-wide copy number variation analysis identifies novel candidate loci associated with pediatric obesity. Eur J Hum Genet 2018; 26:1588-1596. [PMID: 29976977 PMCID: PMC6189095 DOI: 10.1038/s41431-018-0189-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 04/10/2018] [Accepted: 05/15/2018] [Indexed: 12/23/2022] Open
Abstract
Obesity is a multifactorial condition that is highly heritable. There have been ~60 susceptibility loci identified, but they only account for a fraction of cases. As copy number variations (CNVs) have been implicated in the etiology of a multitude of human disorders including obesity, here, we investigated the contribution of rare (<1% population frequency) CNVs in pediatric cases of obesity. We genotyped 67 such individuals, including 22 with co-morbid developmental delay and prioritized rare CNVs at known obesity-associated loci, as well as, those impacting genes involved in energy homeostasis or related processes. We identified clinically relevant or potentially clinically relevant CNVs in 15% (10/67) of individuals. Of these, 4% (3/67) had 16p11.2 microdeletions encompassing the known obesity risk gene SH2B1. Notably, we identified two unrelated probands harboring different 6p22.2 microduplications encompassing SCGN, a potential novel candidate gene for obesity. Further, we identified other biologically relevant candidate genes for pediatric obesity including ARID5B, GPR39, PTPRN2, and HNF4G. We found previously reported candidate loci for obesity, and new ones, suggesting CNV analysis may assist in the diagnosis of pediatric obesity.
Collapse
Affiliation(s)
- Thanuja Selvanayagam
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Susan Walker
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Matthew J Gazzellone
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Barbara Kellam
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Cheryl Cytrynbaum
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Division of Clinical & Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Dimitri J Stavropoulos
- Department of Pediatric Laboratory Medicine, Genome Diagnostics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ping Li
- Division of Endocrinology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Catherine S Birken
- Division of Pediatric Medicine, Hospital for Sick Children, Toronto, ON, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Jill Hamilton
- Division of Endocrinology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Rosanna Weksberg
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.
- Division of Clinical & Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada.
| | - Stephen W Scherer
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- McLaughlin Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
102
|
Dewey D. What Is Comorbidity and Why Does It Matter in Neurodevelopmental Disorders? CURRENT DEVELOPMENTAL DISORDERS REPORTS 2018. [DOI: 10.1007/s40474-018-0152-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
103
|
ASTN2 modulates synaptic strength by trafficking and degradation of surface proteins. Proc Natl Acad Sci U S A 2018; 115:E9717-E9726. [PMID: 30242134 PMCID: PMC6187130 DOI: 10.1073/pnas.1809382115] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Neurogenetic studies demonstrate that copy number variations (CNVs) in the ASTN2 gene occur in patients with neurodevelopmental disorders (NDDs), including autism spectrum. Here, we show that ASTN2 associates with recycling and degradative vesicles in cerebellar neurons, and binds to and promotes the endocytic trafficking and degradation of synaptic proteins. Overexpression of ASTN2 in neurons increases synaptic activity and reduces the levels of ASTN2 binding partners, an effect dependent on its FNIII domain, which is recurrently perturbed by CNVs in patients with NDDs. These findings suggest that ASTN2 is a key regulator of dynamic trafficking of synaptic proteins and lend support to the idea that aberrant regulation of protein homeostasis in neurons is a contributing cause of complex NDDs. Surface protein dynamics dictate synaptic connectivity and function in neuronal circuits. ASTN2, a gene disrupted by copy number variations (CNVs) in neurodevelopmental disorders, including autism spectrum, was previously shown to regulate the surface expression of ASTN1 in glial-guided neuronal migration. Here, we demonstrate that ASTN2 binds to and regulates the surface expression of multiple synaptic proteins in postmigratory neurons by endocytosis, resulting in modulation of synaptic activity. In cerebellar Purkinje cells (PCs), by immunogold electron microscopy, ASTN2 localizes primarily to endocytic and autophagocytic vesicles in the cell soma and in subsets of dendritic spines. Overexpression of ASTN2 in PCs, but not of ASTN2 lacking the FNIII domain, recurrently disrupted by CNVs in patients, including in a family presented here, increases inhibitory and excitatory postsynaptic activity and reduces levels of ASTN2 binding partners. Our data suggest a fundamental role for ASTN2 in dynamic regulation of surface proteins by endocytic trafficking and protein degradation.
Collapse
|
104
|
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: 1.9] [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
|
105
|
Chang Y, Hee S, Lee W, Li H, Chang T, Lin M, Hung Y, Lee I, Hung K, Assimes T, Knowles JW, Nong J, Lee P, Chiu Y, Chuang L. Genome-wide scan for circulating vascular adhesion protein-1 levels: MACROD2 as a potential transcriptional regulator of adipogenesis. J Diabetes Investig 2018; 9:1067-1074. [PMID: 29364582 PMCID: PMC6123039 DOI: 10.1111/jdi.12805] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/10/2018] [Accepted: 01/15/2018] [Indexed: 12/28/2022] Open
Abstract
AIMS/INTRODUCTION Vascular adhesion protein-1 (VAP-1) is a membrane-bound amine oxidase highly expressed in mature adipocytes and released into the circulation. VAP-1 has been strongly implicated in several pathological processes, including diabetes, inflammation, hypertension, hepatic steatosis and renal diseases, and is an important disease marker and therapeutic target. Here, we aimed to identify the genetic loci for circulating VAP-1 levels. MATERIALS AND METHODS We carried out a genomic-wide linkage scan for the quantitative trait locus of circulating VAP-1 levels in 1,100 Han Chinese individuals from 398 families in the Stanford Asian Pacific Program for Hypertension and Insulin Resistance study. Regional association fine mapping was carried out using additional single-nucleotide polymorphisms. RESULTS The estimated heritability of circulating VAP-1 levels is high (h2 = 69%). The most significant quantitative trait locus for circulating VAP-1 was located at 38 cM on chromosome 20, with a maximum empirical logarithm of odds score of 4.11 (P = 6.86 × 10-6 ) in females. Regional single-nucleotide polymorphism fine mapping within a 1-unit support region showed the strongest association signals in the MACRO domain containing 2 (MACROD2) gene in females (P = 5.38 × 10-6 ). Knockdown of MACROD2 significantly suppressed VAP-1 expression in human adipocytes, as well as the expression of key adipogenic genes. Furthermore, MACROD2 expression was found to be positively associated with VAP-1 in human visceral adipose tissue. CONCLUSION MACROD2 is a potential genetic determinant of serum VAP-1 levels, probably through transcriptional regulation of adipogenesis.
Collapse
Affiliation(s)
- Yi‐Cheng Chang
- Graduate Institute of Medical Genomics and ProteomicsCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
- Institute of Biomedical ScienceAcademia SinicaTaipeiTaiwan
- Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Siow‐Wey Hee
- Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Wei‐Jei Lee
- Department of SurgeryMin‐Sheng General HospitalTaoyuanTaiwan
| | - Hung‐Yuan Li
- Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Tien‐Jyun Chang
- Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | | | - Yi‐Jen Hung
- Division of Endocrinology & MetabolismTri‐Service General HospitalNational Defense Medical CenterTaipeiTaiwan
| | - I‐Te Lee
- Department of Internal MedicineDivision of Endocrinology and MetabolismTaichung Veterans General HospitalTaichungTaiwan
| | - Kuan‐Yi Hung
- Institute of Population Health SciencesNational Health Research InstitutesZhunan, MiaoliTaiwan
| | - Themistocles Assimes
- Division of Cardiovascular Medicine and Cardiovascular InstituteDepartment of MedicineStanford University StanfordStanfordCaliforniaUSA
| | - Joshua W Knowles
- Division of Cardiovascular Medicine and Cardiovascular InstituteDepartment of MedicineStanford University StanfordStanfordCaliforniaUSA
| | - Jiun‐Yi Nong
- Graduate Institute of Molecular MedicineCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Po‐Chu Lee
- Department of General SurgeryNational Taiwan University HospitalTaipeiTaiwan
| | - Yen‐Feng Chiu
- Institute of Population Health SciencesNational Health Research InstitutesZhunan, MiaoliTaiwan
| | - Lee‐Ming Chuang
- Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
- Graduate Institute of Molecular MedicineCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
- Graduate Institute of Clinical MedicineCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
- Graduate of Epidemiology and Preventive MedicineCollege of Public HealthNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
106
|
Martin J, Taylor MJ, Lichtenstein P. Assessing the evidence for shared genetic risks across psychiatric disorders and traits. Psychol Med 2018; 48:1759-1774. [PMID: 29198204 PMCID: PMC6088770 DOI: 10.1017/s0033291717003440] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 12/21/2022]
Abstract
Genetic influences play a significant role in risk for psychiatric disorders, prompting numerous endeavors to further understand their underlying genetic architecture. In this paper, we summarize and review evidence from traditional twin studies and more recent genome-wide molecular genetic analyses regarding two important issues that have proven particularly informative for psychiatric genetic research. First, emerging results are beginning to suggest that genetic risk factors for some (but not all) clinically diagnosed psychiatric disorders or extreme manifestations of psychiatric traits in the population share genetic risks with quantitative variation in milder traits of the same disorder throughout the general population. Second, there is now evidence for substantial sharing of genetic risks across different psychiatric disorders. This extends to the level of characteristic traits throughout the population, with which some clinical disorders also share genetic risks. In this review, we summarize and evaluate the evidence for these two issues, for a range of psychiatric disorders. We then critically appraise putative interpretations regarding the potential meaning of genetic correlation across psychiatric phenotypes. We highlight several new methods and studies which are already using these insights into the genetic architecture of psychiatric disorders to gain additional understanding regarding the underlying biology of these disorders. We conclude by outlining opportunities for future research in this area.
Collapse
Affiliation(s)
- Joanna Martin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Mark J. Taylor
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Paul Lichtenstein
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
107
|
Weng X, Liu F, Zhang H, Kan M, Wang T, Dong M, Liu Y. Genome-wide DNA methylation profiling in infants born to gestational diabetes mellitus. Diabetes Res Clin Pract 2018; 142:10-18. [PMID: 29596946 DOI: 10.1016/j.diabres.2018.03.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/01/2018] [Accepted: 03/07/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Offspring exposed to gestational diabetes mellitus (GDM) are at a high risk for metabolic diseases. The mechanisms behind the association between offspring exposed to GDM in utero and an increased risk of health consequences later in life remain unclear. The aim of this study was to clarify the changes in methylation levels in the foetuses of women with GDM and to explore the possible mechanisms linking maternal GDM with a high risk of metabolic diseases in offspring later in life. METHODS A genome-wide comparative methylome analysis on the umbilical cord blood of infants born to 30 women with GDM and 33 women with normal pregnancy was performed using Infinium HumanMethylation 450 BeadChip assays. A quantitative methylation analysis of 18 CpG dinucleotides was verified in the validation umbilical cord blood samples from 102 newborns exposed to GDM and 103 newborns who experienced normal pregnancy by MassARRAY EpiTYPER. RESULTS A total of 4485 differentially methylated sites (DMSs), including 2150 hypermethylated sites and 2335 hypomethylated sites, with a mean β-value difference of >0.05, were identified by the 450k array. Good agreement was observed between the massarray validation data and the 450k array data (R2 > 0.99; P < 0.0001). Thirty-seven CpGs (representing 20 genes) with a β-value difference of > 0.15 between the GDM and healthy groups were identified and showed potential as clinical biomarkers for GDM. "hsa04940: Type I diabetes mellitus" was the most significant Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, with a P-value = 3.20E-07 and 1.36E-02 in the hypermethylated and hypomethylated genepathway enrichment analyses, respectively. In the Gene Ontology (GO) pathway analyses, immune MHC (major histocompatibility complex)-related pathways and neuron development-related pathways were significantly enriched. CONCLUSIONS Our results suggest that GDM has epigenetic effects on genes that are preferentially involved in the Type I diabetes mellitus pathway, immune MHC-related pathways and neuron development-related pathways, with consequences on fetal growth and development, and provide supportive evidence that DNA methylation is involved in fetal metabolic programming.
Collapse
Affiliation(s)
- Xiaoling Weng
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, PR China; Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, PR China
| | - Fatao Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hong Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, PR China
| | - Mengyuan Kan
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Ting Wang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Minyue Dong
- Women's Hospital, School of Medicine, Zhejiang University, PR China
| | - Yun Liu
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, PR China; Key Laboratory of Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, PR China.
| |
Collapse
|
108
|
Ming X, Chen N, Ray C, Brewer G, Kornitzer J, Steer RA. A Gut Feeling: A Hypothesis of the Role of the Microbiome in Attention-Deficit/Hyperactivity Disorders. Child Neurol Open 2018; 5:2329048X18786799. [PMID: 30023407 PMCID: PMC6047248 DOI: 10.1177/2329048x18786799] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 05/25/2018] [Accepted: 06/12/2018] [Indexed: 01/14/2023] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurologic disorder characterized by hyperactivity/impulsivity and/or inattentiveness, with genetic and environmental factors contributing to the disorder. With the growing recognition of the microbiome’s role in many neurological disorders, the authors propose that it may also be implicated in ADHD. Here, we describe several evolving areas of research to support this hypothesis. First, a unique composition of gut bacteria has been identified and linked to behaviors in ADHD. Second, our research found an increased incidence of 2 gastrointestinal symptoms (constipation and flatulence) in children with ADHD, as compared to controls. Finally, emerging data may be interpreted to suggest that immune dysregulation in ADHD be associated with an altered microbiome, low-grade inflammation, and gastrointestinal dysfunction. Although more studies are needed to elucidate exact mechanisms and causality, we propose that an altered microbiome, gastrointestinal symptoms, and immune dysregulation may be associated with the ADHD phenotypes.
Collapse
Affiliation(s)
- Xue Ming
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Neil Chen
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Carly Ray
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Gretchen Brewer
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jeffrey Kornitzer
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Robert A Steer
- Department of Surgery, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| |
Collapse
|
109
|
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: 4.0] [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]
|
110
|
Kebir O, Chaumette B, Krebs MO. Epigenetic variability in conversion to psychosis: novel findings from an innovative longitudinal methylomic analysis. Transl Psychiatry 2018; 8:93. [PMID: 29695761 PMCID: PMC5916914 DOI: 10.1038/s41398-018-0138-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 01/10/2018] [Accepted: 01/31/2018] [Indexed: 01/25/2023] Open
Abstract
Conversion to psychosis is a longitudinal process during which several epigenetic changes have been described. We tested the hypothesis that epigenetic variability in the methylomes of ultra-high risk (UHR) individuals may contribute to the risk of conversion. We studied a longitudinal cohort of UHR individuals (n = 39) and compared two groups (converters, n = 14 vs. non-converters, n = 25). A longitudinal methylomic study was conducted using Infinium HumanMethylation450 BeadChip covering half a million cytosine-phosphate-guanine (CpG) sites across the human genome from whole-blood samples. We used two statistical methods to investigate the variability of methylation probes. (i) The search for longitudinal variable methylation probes (VMPs) based on median comparisons identified two VMPs in converters only. The first CpG was located in the MACROD2 gene and the second CpG was in an intergenic region at 8q24.21. (ii) The detection of outliers using variance analysis related to private epimutations identified a dozen CpGs in converters only and highlighted two genes (RAC1 and SPHK1) from the sphingolipid signaling pathway. Our study is the first to support increased methylome variability during conversion to psychosis. We speculate that stochastic factors could increase DNA methylation variability and have a role in the complex pathophysiology of conversion to psychosis as well as in other psychiatric diseases.
Collapse
Affiliation(s)
- Oussama Kebir
- Centre de Psychiatrie et Neurosciences, Université Paris Descartes, PRES Université Paris Sorbonne Paris Cité, UMR S 894, Paris, France. .,Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, INSERM, UMR S 894, Paris, France. .,CNRS, GDR3557-Institut de Psychiatrie, Paris, France. .,Faculté de Médecine Paris Descartes, Centre Hospitalier Sainte-Annes, Service d'Addictologie «Moreau de Tours», Paris, France.
| | - Boris Chaumette
- 0000 0004 1788 6194grid.469994.fCentre de Psychiatrie et Neurosciences, Université Paris Descartes, PRES Université Paris Sorbonne Paris Cité, UMR S 894 Paris, France ,0000000121866389grid.7429.8Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, INSERM, UMR S 894 Paris, France ,CNRS, GDR3557-Institut de Psychiatrie, Paris, France
| | - Marie-Odile Krebs
- 0000 0004 1788 6194grid.469994.fCentre de Psychiatrie et Neurosciences, Université Paris Descartes, PRES Université Paris Sorbonne Paris Cité, UMR S 894 Paris, France ,0000000121866389grid.7429.8Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, INSERM, UMR S 894 Paris, France ,CNRS, GDR3557-Institut de Psychiatrie, Paris, France ,0000 0001 2200 9055grid.414435.3Faculté de Médecine Paris Descartes, Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| |
Collapse
|
111
|
Ribeiro LF, Verpoort B, de Wit J. Trafficking mechanisms of synaptogenic cell adhesion molecules. Mol Cell Neurosci 2018; 91:34-47. [PMID: 29631018 DOI: 10.1016/j.mcn.2018.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 01/01/2023] Open
Abstract
Nearly every aspect of neuronal function, from wiring to information processing, critically depends on the highly polarized architecture of neurons. Establishing and maintaining the distinct molecular composition of axonal and dendritic compartments requires precise control over the trafficking of the proteins that make up these cellular domains. Synaptic cell adhesion molecules (CAMs), membrane proteins with a critical role in the formation, differentiation and plasticity of synapses, require targeting to the correct pre- or postsynaptic compartment for proper functioning of neural circuits. However, the mechanisms that control the polarized trafficking, synaptic targeting, and synaptic abundance of CAMs are poorly understood. Here, we summarize current knowledge about the sequential trafficking events along the secretory pathway that control the polarized surface distribution of synaptic CAMs, and discuss how their synaptic targeting and abundance is additionally influenced by post-secretory determinants. The identification of trafficking-impairing mutations in CAMs associated with various neurodevelopmental disorders underscores the importance of correct protein trafficking for normal brain function.
Collapse
Affiliation(s)
- Luís F Ribeiro
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Herestraat 49, 3000 Leuven, Belgium
| | - Ben Verpoort
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Herestraat 49, 3000 Leuven, Belgium
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
112
|
Zarrei M, Hicks GG, Reynolds JN, Thiruvahindrapuram B, Engchuan W, Pind M, Lamoureux S, Wei J, Wang Z, Marshall CR, Wintle RF, Chudley AE, Scherer SW. Copy number variation in fetal alcohol spectrum disorder. Biochem Cell Biol 2018. [PMID: 29533680 DOI: 10.1139/bcb-2017-0241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is characterized by a combination of neurological, developmental, and congenital defects that may occur as a consequence of prenatal alcohol exposure. Earlier reports showed that large chromosomal anomalies may link to FASD. Here, we examined the prevalence and types of copy number variations (CNVs) in FASD cases previously diagnosed by a multidisciplinary FASD team in sites across Canada. We genotyped 95 children with FASD and 87 age-matched, typically developing controls on the Illumina Human Omni2.5 SNP (single nucleotide polymorphisms) array platform. We compared their CNVs with those of 10 851 population controls to identify rare CNVs (<0.1% frequency), which may include large unbalanced chromosomal abnormalities, that might be relevant to FASD. In 12/95 (13%) of the FASD cases, rare CNVs were found that impact potentially clinically relevant developmental genes, including the CACNA1H involved in epilepsy and autism, the 3q29 deletion disorder, and others. Our results show that a subset of children diagnosed with FASD have chromosomal deletions and duplications that may co-occur or explain the neurodevelopmental impairments in a diagnosed cohort of FASD individuals. Children suspected to have FASD with or without sentinel facial features of fetal alcohol syndrome and neurodevelopmental delays should potentially be evaluated by a clinical geneticist and possibly have genetic investigations as appropriate to exclude other etiologies.
Collapse
Affiliation(s)
- Mehdi Zarrei
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Geoffrey G Hicks
- b Regenerative Medicine Program, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - James N Reynolds
- c Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada.,d Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Bhooma Thiruvahindrapuram
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Worrawat Engchuan
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Molly Pind
- b Regenerative Medicine Program, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Sylvia Lamoureux
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - John Wei
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Zhouzhi Wang
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Christian R Marshall
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Richard F Wintle
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Albert E Chudley
- e Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.,f Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Stephen W Scherer
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada.,g Department of Molecular Genetics and McLaughlin Centre, University of Toronto, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
113
|
Bonvicini C, Faraone SV, Scassellati C. Common and specific genes and peripheral biomarkers in children and adults with attention-deficit/hyperactivity disorder. World J Biol Psychiatry 2018; 19:80-100. [PMID: 28097908 PMCID: PMC5568996 DOI: 10.1080/15622975.2017.1282175] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Elucidating the biological mechanisms involved in attention-deficit/hyperactivity disorder (ADHD) has been challenging. Relatively unexplored is the fact that these mechanisms can differ with age. METHODS We present an overview on the major differences between children and adults with ADHD, describing several studies from genomics to metabolomics performed in ADHD children and in adults (cADHD and aADHD, respectively). A systematic search (up until February 2016) was conducted. RESULTS From a PRISMA flow-chart, a total of 350 and 91 genomics and metabolomics studies were found to be elligible for cADHD and aADHD, respectively. For children, associations were found for genes belonging to dopaminergic (SLC6A3, DRD4 and MAOA) and neurodevelopmental (LPHN3 and DIRAS2) systems and OPRM1 (Yates corrected P = 0.016; OR = 2.27 95%CI: 1.15-4.47). Studies of adults have implicated circadian rhythms genes, HTR2A, MAOB and a more generic neurodevelopmental/neurite outgrowth network (BCHE, SNAP25, BAIAP2, NOS1/NO, KCNIP4 and SPOCK3; Yates corrected P = 0.007; OR = 3.30 95%CI: 1.33-8.29). In common among cADHD and aADHD, the most significant findings are for oxidative stress proteins (MAD, SOD, PON1, ARES, TOS, TAS and OSI), and, in the second level, DISC1, DBH, DDC, microRNA and adiponectin. CONCLUSIONS Through a convergent functional genomics, this review contributes to clarification of which genetic/biological mechanisms differ with age. The effects of some genes do not change throughout the lifetime, whereas others are linked to age-specific stages. Additional research and further studies are needed to generate firmer conclusions that might someday be useful for predicting the remission and persistence of the disorder. Despite the limitations, some of these genes/proteins could be potential useful biomarkers to discriminate cADHD from aADHD.
Collapse
Affiliation(s)
- Cristian Bonvicini
- Genetics Unit, IRCCS “Centro S. Giovanni di Dio” Fatebenefratelli, Brescia, Italy
| | - Stephen V. Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA; K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Catia Scassellati
- Genetics Unit, IRCCS “Centro S. Giovanni di Dio” Fatebenefratelli, Brescia, Italy
| |
Collapse
|
114
|
McCammon JM, Blaker-Lee A, Chen X, Sive H. The 16p11.2 homologs fam57ba and doc2a generate certain brain and body phenotypes. Hum Mol Genet 2018; 26:3699-3712. [PMID: 28934389 PMCID: PMC5886277 DOI: 10.1093/hmg/ddx255] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/29/2017] [Indexed: 01/28/2023] Open
Abstract
Deletion of the 16p11.2 CNV affects 25 core genes and is associated with multiple symptoms affecting brain and body, including seizures, hyperactivity, macrocephaly, and obesity. Available data suggest that most symptoms are controlled by haploinsufficiency of two or more 16p11.2 genes. To identify interacting 16p11.2 genes, we used a pairwise partial loss of function antisense screen for embryonic brain morphology, using the accessible zebrafish model. fam57ba, encoding a ceramide synthase, was identified as interacting with the doc2a gene, encoding a calcium-sensitive exocytosis regulator, a genetic interaction not previously described. Using genetic mutants, we demonstrated that doc2a+/− fam57ba+/− double heterozygotes show hyperactivity and increased seizure susceptibility relative to wild-type or single doc2a−/− or fam57ba−/− mutants. Additionally, doc2a+/− fam57ba+/− double heterozygotes demonstrate the increased body length and head size. Single doc2a+/− and fam57ba+/− heterozygotes do not show a body size increase; however, fam57ba−/− homozygous mutants show a strongly increased head size and body length, suggesting a greater contribution from fam57ba to the haploinsufficient interaction between doc2a and fam57ba. The doc2a+/− fam57ba+/− interaction has not been reported before, nor has any 16p11.2 gene previously been linked to increased body size. These findings demonstrate that one pair of 16p11.2 homologs can regulate both brain and body phenotypes that are reflective of those in people with 16p11.2 deletion. Together, these findings suggest that dysregulation of ceramide pathways and calcium sensitive exocytosis underlies seizures and large body size associated with 16p11.2 homologs in zebrafish. The data inform consideration of mechanisms underlying human 16p11.2 deletion symptoms.
Collapse
Affiliation(s)
| | - Alicia Blaker-Lee
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Xiao Chen
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hazel Sive
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
115
|
Kuppili PP. Commentary. J Neurosci Rural Pract 2017; 8:662-663. [PMID: 29204035 PMCID: PMC5709898 DOI: 10.4103/jnrp.jnrp_408_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Pooja Patnaik Kuppili
- Department of Psychiatry, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| |
Collapse
|
116
|
Gur RE, Bassett AS, McDonald-McGinn DM, Bearden CE, Chow E, Emanuel BS, Owen M, Swillen A, Van den Bree M, Vermeesch J, Vorstman JAS, Warren S, Lehner T, Morrow B, The International 22q11.2 Deletion Syndrome Brain Behavior Consortium. A neurogenetic model for the study of schizophrenia spectrum disorders: the International 22q11.2 Deletion Syndrome Brain Behavior Consortium. Mol Psychiatry 2017; 22:1664-1672. [PMID: 28761081 PMCID: PMC5935262 DOI: 10.1038/mp.2017.161] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/14/2017] [Accepted: 06/19/2017] [Indexed: 02/07/2023]
Abstract
Rare copy number variants contribute significantly to the risk for schizophrenia, with the 22q11.2 locus consistently implicated. Individuals with the 22q11.2 deletion syndrome (22q11DS) have an estimated 25-fold increased risk for schizophrenia spectrum disorders, compared to individuals in the general population. The International 22q11DS Brain Behavior Consortium is examining this highly informative neurogenetic syndrome phenotypically and genomically. Here we detail the procedures of the effort to characterize the neuropsychiatric and neurobehavioral phenotypes associated with 22q11DS, focusing on schizophrenia and subthreshold expression of psychosis. The genomic approach includes a combination of whole-genome sequencing and genome-wide microarray technologies, allowing the investigation of all possible DNA variation and gene pathways influencing the schizophrenia-relevant phenotypic expression. A phenotypically rich data set provides a psychiatrically well-characterized sample of unprecedented size (n=1616) that informs the neurobehavioral developmental course of 22q11DS. This combined set of phenotypic and genomic data will enable hypothesis testing to elucidate the mechanisms underlying the pathogenesis of schizophrenia spectrum disorders.
Collapse
Affiliation(s)
- RE Gur
- Perelman School of Medicine and Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - AS Bassett
- Centre for Addiction and Mental Health, Toronto General Hospital and the University of Toronto, Toronto, ON, Canada
| | - DM McDonald-McGinn
- The Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, USA
| | - CE Bearden
- University of California Los Angeles, Los Angeles, CA, USA
| | - E Chow
- Centre for Addiction and Mental Health, Toronto General Hospital and the University of Toronto, Toronto, ON, Canada
| | - BS Emanuel
- The Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, USA
| | - M Owen
- Cardiff University, Cardiff, UK
| | - A Swillen
- Katholieke University, Leuven, Belgium
| | | | | | - JAS Vorstman
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S Warren
- Emory University, Atlanta, GA, USA
| | - T Lehner
- National Institute of Mental Health, Bethesda, MD, USA
| | - B Morrow
- Albert Einstein College of Medicine, New York, NY, USA
| | | |
Collapse
|
117
|
Meyers JL, Zhang J, Wang JC, Su J, Kuo SI, Kapoor M, Wetherill L, Bertelsen S, Lai D, Salvatore JE, Kamarajan C, Chorlian D, Agrawal A, Almasy L, Bauer L, Bucholz KK, Chan G, Hesselbrock V, Koganti L, Kramer J, Kuperman S, Manz N, Pandey A, Seay M, Scott D, Taylor RE, Dick DM, Edenberg HJ, Goate A, Foroud T, Porjesz B. An endophenotype approach to the genetics of alcohol dependence: a genome wide association study of fast beta EEG in families of African ancestry. Mol Psychiatry 2017; 22:1767-1775. [PMID: 28070124 PMCID: PMC5503794 DOI: 10.1038/mp.2016.239] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/24/2016] [Accepted: 10/27/2016] [Indexed: 01/16/2023]
Abstract
Fast beta (20-28 Hz) electroencephalogram (EEG) oscillatory activity may be a useful endophenotype for studying the genetics of disorders characterized by neural hyperexcitability, including substance use disorders (SUDs). However, the genetic underpinnings of fast beta EEG have not previously been studied in a population of African-American ancestry (AA). In a sample of 2382 AA individuals from 482 families drawn from the Collaborative Study on the Genetics of Alcoholism (COGA), we performed a genome-wide association study (GWAS) on resting-state fast beta EEG power. To further characterize our genetic findings, we examined the functional and clinical/behavioral significance of GWAS variants. Ten correlated single-nucleotide polymorphisms (SNPs) (r2>0.9) located in an intergenic region on chromosome 3q26 were associated with fast beta EEG power at P<5 × 10-8. The most significantly associated SNP, rs11720469 (β: -0.124; P<4.5 × 10-9), is also an expression quantitative trait locus for BCHE (butyrylcholinesterase), expressed in thalamus tissue. Four of the genome-wide SNPs were also associated with Diagnostic and Statistical Manual of Mental Disorders Alcohol Dependence in COGA AA families, and two (rs13093097, rs7428372) were replicated in an independent AA sample (Gelernter et al.). Analyses in the AA adolescent/young adult (offspring from COGA families) subsample indicated association of rs11720469 with heavy episodic drinking (frequency of consuming 5+ drinks within 24 h). Converging findings presented in this study provide support for the role of genetic variants within 3q26 in neural and behavioral disinhibition. These novel genetic findings highlight the importance of including AA populations in genetics research on SUDs and the utility of the endophenotype approach in enhancing our understanding of mechanisms underlying addiction susceptibility.
Collapse
Affiliation(s)
- J L Meyers
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - J Zhang
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - J C Wang
- Department of Neuroscience, Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Su
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - S I Kuo
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - M Kapoor
- Department of Neuroscience, Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - L Wetherill
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S Bertelsen
- Department of Neuroscience, Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J E Salvatore
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - C Kamarajan
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - D Chorlian
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - A Agrawal
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - L Almasy
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - L Bauer
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
| | - K K Bucholz
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - G Chan
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
| | - V Hesselbrock
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
| | - L Koganti
- Department of Neuroscience, Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Kramer
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - S Kuperman
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - N Manz
- Department of Physics, The College of Wooster, Wooster, OH, USA
| | - A Pandey
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - M Seay
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - D Scott
- Collaborative Alcohol Research Center, Howard University College of Medicine, Washington, DC, USA
| | - R E Taylor
- Collaborative Alcohol Research Center, Howard University College of Medicine, Washington, DC, USA
| | - D M Dick
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - H J Edenberg
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Goate
- Department of Neuroscience, Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - T Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - B Porjesz
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| |
Collapse
|
118
|
Lowther C, Merico D, Costain G, Waserman J, Boyd K, Noor A, Speevak M, Stavropoulos DJ, Wei J, Lionel AC, Marshall CR, Scherer SW, Bassett AS. Impact of IQ on the diagnostic yield of chromosomal microarray in a community sample of adults with schizophrenia. Genome Med 2017; 9:105. [PMID: 29187259 PMCID: PMC5708103 DOI: 10.1186/s13073-017-0488-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/01/2017] [Indexed: 11/10/2022] Open
Abstract
Background Schizophrenia is a severe psychiatric disorder associated with IQ deficits. Rare copy number variations (CNVs) have been established to play an important role in the etiology of schizophrenia. Several of the large rare CNVs associated with schizophrenia have been shown to negatively affect IQ in population-based controls where no major neuropsychiatric disorder is reported. The aim of this study was to examine the diagnostic yield of microarray testing and the functional impact of genome-wide rare CNVs in a community ascertained cohort of adults with schizophrenia and low (< 85) or average (≥ 85) IQ. Methods We recruited 546 adults of European ancestry with schizophrenia from six community psychiatric clinics in Canada. Each individual was assigned to the low or average IQ group based on standardized tests and/or educational attainment. We used rigorous methods to detect genome-wide rare CNVs from high-resolution microarray data. We compared the burden of rare CNVs classified as pathogenic or as a variant of unknown significance (VUS) between each of the IQ groups and the genome-wide burden and functional impact of rare CNVs after excluding individuals with a pathogenic CNV. Results There were 39/546 (7.1%; 95% confidence interval [CI] = 5.2–9.7%) schizophrenia participants with at least one pathogenic CNV detected, significantly more of whom were from the low IQ group (odds ratio [OR] = 5.01 [2.28–11.03], p = 0.0001). Secondary analyses revealed that individuals with schizophrenia and average IQ had the lowest yield of pathogenic CNVs (n = 9/325; 2.8%), followed by those with borderline intellectual functioning (n = 9/130; 6.9%), non-verbal learning disability (n = 6/29; 20.7%), and co-morbid intellectual disability (n = 15/62; 24.2%). There was no significant difference in the burden of rare CNVs classified as a VUS between any of the IQ subgroups. There was a significantly (p=0.002) increased burden of rare genic duplications in individuals with schizophrenia and low IQ that persisted after excluding individuals with a pathogenic CNV. Conclusions Using high-resolution microarrays we were able to demonstrate for the first time that the burden of pathogenic CNVs in schizophrenia differs significantly between IQ subgroups. The results of this study have implications for clinical practice and may help inform future rare variant studies of schizophrenia using next-generation sequencing technologies. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0488-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chelsea Lowther
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, Canada, M5S 2S1.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Daniele Merico
- Deep Genomics Inc, Toronto, ON, Canada.,The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Gregory Costain
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, Canada, M5S 2S1.,Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Kerry Boyd
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Abdul Noor
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Marsha Speevak
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | | | - John Wei
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anath C Lionel
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Christian R Marshall
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,McLaughlin Centre, University of Toronto, Toronto, ON, Canada
| | - Anne S Bassett
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, Canada, M5S 2S1. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Toronto General Research Institute, University Health Network, Toronto, ON, Canada. .,Cambell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada. .,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
119
|
Gilbert J, Man HY. Fundamental Elements in Autism: From Neurogenesis and Neurite Growth to Synaptic Plasticity. Front Cell Neurosci 2017; 11:359. [PMID: 29209173 PMCID: PMC5701944 DOI: 10.3389/fncel.2017.00359] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/31/2017] [Indexed: 01/12/2023] Open
Abstract
Autism spectrum disorder (ASD) is a set of neurodevelopmental disorders with a high prevalence and impact on society. ASDs are characterized by deficits in both social behavior and cognitive function. There is a strong genetic basis underlying ASDs that is highly heterogeneous; however, multiple studies have highlighted the involvement of key processes, including neurogenesis, neurite growth, synaptogenesis and synaptic plasticity in the pathophysiology of neurodevelopmental disorders. In this review article, we focus on the major genes and signaling pathways implicated in ASD and discuss the cellular, molecular and functional studies that have shed light on common dysregulated pathways using in vitro, in vivo and human evidence. HighlightsAutism spectrum disorder (ASD) has a prevalence of 1 in 68 children in the United States. ASDs are highly heterogeneous in their genetic basis. ASDs share common features at the cellular and molecular levels in the brain. Most ASD genes are implicated in neurogenesis, structural maturation, synaptogenesis and function.
Collapse
Affiliation(s)
- James Gilbert
- Department of Biology, Boston University, Boston, MA, United States
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, United States.,Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
120
|
Forero A, Rivero O, Wäldchen S, Ku HP, Kiser DP, Gärtner Y, Pennington LS, Waider J, Gaspar P, Jansch C, Edenhofer F, Resink TJ, Blum R, Sauer M, Lesch KP. Cadherin-13 Deficiency Increases Dorsal Raphe 5-HT Neuron Density and Prefrontal Cortex Innervation in the Mouse Brain. Front Cell Neurosci 2017; 11:307. [PMID: 29018333 PMCID: PMC5623013 DOI: 10.3389/fncel.2017.00307] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 09/15/2017] [Indexed: 01/29/2023] Open
Abstract
Background: During early prenatal stages of brain development, serotonin (5-HT)-specific neurons migrate through somal translocation to form the raphe nuclei and subsequently begin to project to their target regions. The rostral cluster of cells, comprising the median and dorsal raphe (DR), innervates anterior regions of the brain, including the prefrontal cortex. Differential analysis of the mouse 5-HT system transcriptome identified enrichment of cell adhesion molecules in 5-HT neurons of the DR. One of these molecules, cadherin-13 (Cdh13) has been shown to play a role in cell migration, axon pathfinding, and synaptogenesis. This study aimed to investigate the contribution of Cdh13 to the development of the murine brain 5-HT system. Methods: For detection of Cdh13 and components of the 5-HT system at different embryonic developmental stages of the mouse brain, we employed immunofluorescence protocols and imaging techniques, including epifluorescence, confocal and structured illumination microscopy. The consequence of CDH13 loss-of-function mutations on brain 5-HT system development was explored in a mouse model of Cdh13 deficiency. Results: Our data show that in murine embryonic brain Cdh13 is strongly expressed on 5-HT specific neurons of the DR and in radial glial cells (RGCs), which are critically involved in regulation of neuronal migration. We observed that 5-HT neurons are intertwined with these RGCs, suggesting that these neurons undergo RGC-guided migration. Cdh13 is present at points of intersection between these two cell types. Compared to wildtype controls, Cdh13-deficient mice display increased cell densities in the DR at embryonic stages E13.5, E17.5, and adulthood, and higher serotonergic innervation of the prefrontal cortex at E17.5. Conclusion: Our findings provide evidence for a role of CDH13 in the development of the serotonergic system in early embryonic stages. Specifically, we indicate that Cdh13 deficiency affects the cell density of the developing DR and the posterior innervation of the prefrontal cortex (PFC), and therefore might be involved in the migration, axonal outgrowth and terminal target finding of DR 5-HT neurons. Dysregulation of CDH13 expression may thus contribute to alterations in this system of neurotransmission, impacting cognitive function, which is frequently impaired in neurodevelopmental disorders including attention-deficit/hyperactivity and autism spectrum disorders.
Collapse
Affiliation(s)
- Andrea Forero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Olga Rivero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Sina Wäldchen
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Hsing-Ping Ku
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Dominik P Kiser
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Yvonne Gärtner
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Laura S Pennington
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Jonas Waider
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Patricia Gaspar
- Institut du Fer á Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839, Paris, France
| | - Charline Jansch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Frank Edenhofer
- Department of Genomics, Stem Cell Biology and Regenerative Medicine, Institute of Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens-University Innsbruck, Innsbruck, Austria.,Stem Cell Biology and Regenerative Medicine Group, Institute of Anatomy and Cell Biology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Thérèse J Resink
- Laboratory for Signal Transduction, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Robert Blum
- Department of Clinical Neurobiology, University of Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - 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 of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
121
|
Lowther C, Costain G, Baribeau DA, Bassett AS. Genomic Disorders in Psychiatry-What Does the Clinician Need to Know? Curr Psychiatry Rep 2017; 19:82. [PMID: 28929285 DOI: 10.1007/s11920-017-0831-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the role of genomic disorders in various psychiatric conditions and to highlight important recent advances in the field that are of potential clinical relevance. RECENT FINDINGS Genomic disorders are caused by large rare recurrent deletions and duplications at certain chromosomal "hotspots" (e.g., 22q11.2, 16p11.2, 15q11-q13, 1q21.1, 15q13.3) across the genome. Most overlap multiple genes, affect development, and are associated with variable cognitive and other neuropsychiatric expression. Although individually rare, genomic disorders collectively account for a significant minority of intellectual disability, autism spectrum disorder, and schizophrenia. Genome-wide chromosomal microarray analysis is capable of detecting all genomic disorders in a single test, offering the first opportunity for routine clinical genetic testing in psychiatric practice.
Collapse
Affiliation(s)
- Chelsea Lowther
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, M5S 2S1, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Gregory Costain
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, and Medical Genetics Residency Training Program, University of Toronto, Toronto, ON, Canada
| | | | - Anne S Bassett
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, M5S 2S1, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Department of Psychiatry, University of Toronto, Toronto, ON, Canada. .,Dalglish Family 22q Clinic for Adults with 22q11.2 Deletion Syndrome and Toronto General Research Institute, University Health Network, and Campbell Family Mental Health Research Institute, Toronto, ON, Canada.
| |
Collapse
|
122
|
Klein M, Onnink M, van Donkelaar M, Wolfers T, Harich B, Shi Y, Dammers J, Arias-Vásquez A, Hoogman M, Franke B. Brain imaging genetics in ADHD and beyond - Mapping pathways from gene to disorder at different levels of complexity. Neurosci Biobehav Rev 2017; 80:115-155. [PMID: 28159610 PMCID: PMC6947924 DOI: 10.1016/j.neubiorev.2017.01.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 12/08/2016] [Accepted: 01/09/2017] [Indexed: 01/03/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a common and often persistent neurodevelopmental disorder. Beyond gene-finding, neurobiological parameters, such as brain structure, connectivity, and function, have been used to link genetic variation to ADHD symptomatology. We performed a systematic review of brain imaging genetics studies involving 62 ADHD candidate genes in childhood and adult ADHD cohorts. Fifty-one eligible research articles described studies of 13 ADHD candidate genes. Almost exclusively, single genetic variants were studied, mostly focussing on dopamine-related genes. While promising results have been reported, imaging genetics studies are thus far hampered by methodological differences in study design and analysis methodology, as well as limited sample sizes. Beyond reviewing imaging genetics studies, we also discuss the need for complementary approaches at multiple levels of biological complexity and emphasize the importance of combining and integrating findings across levels for a better understanding of biological pathways from gene to disease. These may include multi-modal imaging genetics studies, bioinformatic analyses, and functional analyses of cell and animal models.
Collapse
Affiliation(s)
- Marieke Klein
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Marten Onnink
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Marjolein van Donkelaar
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Thomas Wolfers
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Benjamin Harich
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Yan Shi
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Janneke Dammers
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands; Department of Psychiatry, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Alejandro Arias-Vásquez
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands; Department of Psychiatry, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands; Department of Cognitive Neuroscience, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Martine Hoogman
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Barbara Franke
- Department of Human Genetics, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands; Department of Psychiatry, Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| |
Collapse
|
123
|
Abstract
Human genetic studies have been the driving force in bringing to light the underlying biology of psychiatric conditions. As these studies fill in the gaps in our knowledge of the mechanisms at play, we will be better equipped to design therapies in rational and targeted ways, or repurpose existing therapies in previously unanticipated ways. This review is intended for those unfamiliar with psychiatric genetics as a field and provides a primer on different modes of genetic variation, the technologies currently used to probe them, and concepts that provide context for interpreting the gene-phenotype relationship. Like other subfields in human genetics, psychiatric genetics is moving from microarray technology to sequencing-based approaches as barriers of cost and expertise are removed, and the ramifications of this transition are discussed here. A summary is then given of recent genetic discoveries in a number of neuropsychiatric conditions, with particular emphasis on neurodevelopmental conditions. The general impact of genetics on drug development has been to underscore the extensive etiological heterogeneity in seemingly cohesive diagnostic categories. Consequently, the path forward is not in therapies hoping to reach large swaths of patients sharing a clinically defined diagnosis, but rather in targeting patients belonging to specific "biotypes" defined through a combination of objective, quantifiable data, including genotype.
Collapse
Affiliation(s)
- Jacob J Michaelson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
- Department of Biomedical Engineering, University of Iowa College of Engineering, Iowa City, IA, USA.
- Department of Communication Sciences and Disorders, University of Iowa College of Liberal Arts and Sciences, Iowa City, IA, USA.
- Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA, USA.
- Genetics Cluster Initiative, University of Iowa, Iowa City, IA, USA.
- The DeLTA Center, University of Iowa, Iowa City, IA, USA.
- University of Iowa Informatics Initiative, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
124
|
Watanabe M, Hatakeyama S. TRIM proteins and diseases. J Biochem 2017; 161:135-144. [PMID: 28069866 DOI: 10.1093/jb/mvw087] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 09/20/2016] [Indexed: 12/20/2022] Open
Abstract
Ubiquitination is one of the posttranslational modifications that regulates a number of intracellular events including signal transduction, protein quality control, transcription, cell cycle, apoptosis and development. The ubiquitin system functions as a garbage machine to degrade target proteins and as a regulator for several signalling pathways. Biochemical reaction of ubiquitination requires several enzymes including E1, E2 and E3, and E3 ubiquitin ligases play roles as receptors for recognizing target proteins. Most of the tripartite motif (TRIM) proteins are E3 ubiquitin ligases. Recent studies have shown that some TRIM proteins function as important regulators for a variety of diseases including cancer, inflammatory diseases, infectious diseases, neuropsychiatric disorders, chromosomal abnormalities and developmental diseases. In this review, we summarize the involvement of TRIM proteins in the aetiology of various diseases.
Collapse
Affiliation(s)
- Masashi Watanabe
- Department of Biochemistry, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Shigetsugu Hatakeyama
- Department of Biochemistry, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| |
Collapse
|
125
|
Berkowicz SR, Giousoh A, Bird PI. Neurodevelopmental MACPFs: The vertebrate astrotactins and BRINPs. Semin Cell Dev Biol 2017; 72:171-181. [PMID: 28506896 DOI: 10.1016/j.semcdb.2017.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 04/27/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023]
Abstract
Astrotactins (ASTNs) and Bone morphogenetic protein/retinoic acid inducible neural-specific proteins (BRINPs) are two groups of Membrane Attack Complex/Perforin (MACPF) superfamily proteins that show overlapping expression in the developing and mature vertebrate nervous system. ASTN(1-2) and BRINP(1-3) genes are found at conserved loci in humans that have been implicated in neurodevelopmental disorders (NDDs). Here we review the tissue distribution and cellular localization of these proteins, and discuss recent studies that provide insight into their structure and interactions. We highlight the genetic relationships and co-expression of Brinps and Astns; and review recent knock-out mouse phenotypes that indicate a possible overlap in protein function between ASTNs and BRINPs.
Collapse
Affiliation(s)
- Susan R Berkowicz
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, 3800, Australia.
| | - Aminah Giousoh
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, 3800, Australia
| | - Phillip I Bird
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, 3800, Australia
| |
Collapse
|
126
|
Srivastav S, Walitza S, Grünblatt E. Emerging role of miRNA in attention deficit hyperactivity disorder: a systematic review. ACTA ACUST UNITED AC 2017; 10:49-63. [DOI: 10.1007/s12402-017-0232-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/29/2017] [Indexed: 12/11/2022]
|
127
|
Kleijer KTE, van Nieuwenhuize D, Spierenburg HA, Gregorio-Jordan S, Kas MJH, Burbach JPH. Structural abnormalities in the primary somatosensory cortex and a normal behavioral profile in Contactin-5 deficient mice. Cell Adh Migr 2017; 12:5-18. [PMID: 28346043 PMCID: PMC5810773 DOI: 10.1080/19336918.2017.1288788] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Contactin-5 (Cntn5) is an immunoglobulin cell adhesion molecule that is exclusively expressed in the central nervous system. In view of its association with neurodevelopmental disorders, particularly autism spectrum disorder (ASD), this study focused on Cntn5-positive areas in the forebrain and aimed to explore the morphological and behavioral phenotypes of the Cntn5 null mutant (Cntn5−/−) mouse in relation to these areas and ASD symptomatology. A newly generated antibody enabled us to elaborately describe the spatial expression pattern of Cntn5 in P7 wild type (Cntn5+/+) mice. The Cntn5 expression pattern included strong expression in the cerebral cortex, hippocampus and mammillary bodies in addition to described previously brain nuclei of the auditory pathway and the dorsal thalamus. Thinning of the primary somatosensory (S1) cortex was found in Cntn5−/− mice and ascribed to a misplacement of Cntn5-ablated cells. This phenotype was accompanied by a reduction in the barrel/septa ratio of the S1 barrel field. The structure and morphology of the hippocampus was intact in Cntn5−/− mice. A set of behavioral experiments including social, exploratory and repetitive behaviors showed that these were unaffected in Cntn5−/− mice. Taken together, these data demonstrate a selective role of Cntn5 in development of the cerebral cortex without overt behavioral phenotypes.
Collapse
Affiliation(s)
- Kristel T E Kleijer
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| | - Denise van Nieuwenhuize
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| | - Henk A Spierenburg
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| | - Sara Gregorio-Jordan
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| | - Martien J H Kas
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| | - J Peter H Burbach
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| |
Collapse
|
128
|
Collins RL, Brand H, Redin CE, Hanscom C, Antolik C, Stone MR, Glessner JT, Mason T, Pregno G, Dorrani N, Mandrile G, Giachino D, Perrin D, Walsh C, Cipicchio M, Costello M, Stortchevoi A, An JY, Currall BB, Seabra CM, Ragavendran A, Margolin L, Martinez-Agosto JA, Lucente D, Levy B, Sanders SJ, Wapner RJ, Quintero-Rivera F, Kloosterman W, Talkowski ME. Defining the diverse spectrum of inversions, complex structural variation, and chromothripsis in the morbid human genome. Genome Biol 2017; 18:36. [PMID: 28260531 PMCID: PMC5338099 DOI: 10.1186/s13059-017-1158-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/20/2017] [Indexed: 12/13/2022] Open
Abstract
Background Structural variation (SV) influences genome organization and contributes to human disease. However, the complete mutational spectrum of SV has not been routinely captured in disease association studies. Results We sequenced 689 participants with autism spectrum disorder (ASD) and other developmental abnormalities to construct a genome-wide map of large SV. Using long-insert jumping libraries at 105X mean physical coverage and linked-read whole-genome sequencing from 10X Genomics, we document seven major SV classes at ~5 kb SV resolution. Our results encompass 11,735 distinct large SV sites, 38.1% of which are novel and 16.8% of which are balanced or complex. We characterize 16 recurrent subclasses of complex SV (cxSV), revealing that: (1) cxSV are larger and rarer than canonical SV; (2) each genome harbors 14 large cxSV on average; (3) 84.4% of large cxSVs involve inversion; and (4) most large cxSV (93.8%) have not been delineated in previous studies. Rare SVs are more likely to disrupt coding and regulatory non-coding loci, particularly when truncating constrained and disease-associated genes. We also identify multiple cases of catastrophic chromosomal rearrangements known as chromoanagenesis, including somatic chromoanasynthesis, and extreme balanced germline chromothripsis events involving up to 65 breakpoints and 60.6 Mb across four chromosomes, further defining rare categories of extreme cxSV. Conclusions These data provide a foundational map of large SV in the morbid human genome and demonstrate a previously underappreciated abundance and diversity of cxSV that should be considered in genomic studies of human disease. Electronic supplementary material The online version of this article (doi:10.1186/s13059-017-1158-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ryan L Collins
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Bioinformatics and Integrative Genomics, Division of Medical Sciences, Harvard Medical School, Boston, MA, 02115, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Harrison Brand
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Claire E Redin
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Carrie Hanscom
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Caroline Antolik
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Matthew R Stone
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Joseph T Glessner
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Tamara Mason
- Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Giulia Pregno
- Medical Genetics Unit, Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Naghmeh Dorrani
- Department of Pathology & Laboratory Medicine and UCLA Clinical Genomics Center, David Geffen School of Medicine, University of California Los Angeles, UCLA, Los Angeles, CA, 90095, USA
| | - Giorgia Mandrile
- Medical Genetics Unit, Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Daniela Giachino
- Medical Genetics Unit, Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Danielle Perrin
- Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Cole Walsh
- Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Michelle Cipicchio
- Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Maura Costello
- Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Alexei Stortchevoi
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Joon-Yong An
- Department of Psychiatry, University of California San Francisco, San Francisco, CA, 94103, USA
| | - Benjamin B Currall
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Catarina M Seabra
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA.,GABBA Program, University of Porto, Porto, 4099-002, Portugal
| | - Ashok Ragavendran
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Lauren Margolin
- Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Julian A Martinez-Agosto
- Department of Pathology & Laboratory Medicine and UCLA Clinical Genomics Center, David Geffen School of Medicine, University of California Los Angeles, UCLA, Los Angeles, CA, 90095, USA
| | - Diane Lucente
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Brynn Levy
- Department of Pathology, Columbia University, New York, NY, 10032, USA
| | - Stephan J Sanders
- Department of Psychiatry, University of California San Francisco, San Francisco, CA, 94103, USA
| | - Ronald J Wapner
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Fabiola Quintero-Rivera
- Department of Pathology & Laboratory Medicine and UCLA Clinical Genomics Center, David Geffen School of Medicine, University of California Los Angeles, UCLA, Los Angeles, CA, 90095, USA
| | - Wigard Kloosterman
- Department of Medical Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, 3584CG, The Netherlands
| | - Michael E Talkowski
- Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, and Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA. .,Program in Bioinformatics and Integrative Genomics, Division of Medical Sciences, Harvard Medical School, Boston, MA, 02115, USA. .,Program in Population and Medical Genetics and Genomics Platform, The Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
129
|
A current view on contactin-4, -5, and -6: Implications in neurodevelopmental disorders. Mol Cell Neurosci 2017; 81:72-83. [PMID: 28064060 DOI: 10.1016/j.mcn.2016.12.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/23/2016] [Accepted: 12/25/2016] [Indexed: 12/30/2022] Open
Abstract
Contactins (Cntns) are a six-member subgroup of the immunoglobulin cell adhesion molecule superfamily (IgCAMs) with pronounced brain expression and function. Recent genetic studies of neuropsychiatric disorders have pinpointed contactin-4 (CNTN4), contactin-5 (CNTN5) and contactin-6 (CNTN6) as candidate genes in neurodevelopmental disorders, particularly in autism spectrum disorders (ASDs), but also in intellectual disability, schizophrenia (SCZ), attention-deficit hyperactivity disorder (ADHD), bipolar disorder (BD), alcohol use disorder (AUD) and anorexia nervosa (AN). This suggests that they have important functions during neurodevelopment. This suggestion is supported by data showing that neurite outgrowth, cell survival and neural circuit formation can be affected by disruption of these genes. Here, we review the current genetic data about their involvement in neuropsychiatric disorders and explore studies on how null mutations affect mouse behavior. Finally, we highlight to role of protein-protein interactions in the potential mechanism of action of Cntn4, -5 and -6 and emphasize that complexes with other membrane proteins may play a role in neuronal developmental functions.
Collapse
|
130
|
Gazzellone MJ, Zarrei M, Burton CL, Walker S, Uddin M, Shaheen SM, Coste J, Rajendram R, Schachter RJ, Colasanto M, Hanna GL, Rosenberg DR, Soreni N, Fitzgerald KD, Marshall CR, Buchanan JA, Merico D, Arnold PD, Scherer SW. Uncovering obsessive-compulsive disorder risk genes in a pediatric cohort by high-resolution analysis of copy number variation. J Neurodev Disord 2016; 8:36. [PMID: 27777633 PMCID: PMC5070001 DOI: 10.1186/s11689-016-9170-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 10/04/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Obsessive-compulsive disorder (OCD) is a heterogeneous neuropsychiatric condition, thought to have a significant genetic component. When onset occurs in childhood, affected individuals generally exhibit different characteristics from adult-onset OCD, including higher prevalence in males and increased heritability. Since neuropsychiatric conditions are associated with copy number variations (CNVs), we considered their potential role in the etiology of OCD. METHODS We genotyped 307 unrelated pediatric probands with idiopathic OCD (including 174 that were part of complete parent-child trios) and compared their genotypes with those of 3861 population controls, to identify rare CNVs (<0.5 % frequency) of at least 15 kb in size that might contribute to OCD. RESULTS We uncovered de novo CNVs in 4/174 probands (2.3 %). Our case cohort was enriched for CNVs in genes that encode targets of the fragile X mental retardation protein (nominal p = 1.85 × 10-03; FDR=0.09), similar to previous findings in autism and schizophrenia. These results also identified deletions or duplications of exons in genes involved in neuronal migration (ASTN2), synapse formation (NLGN1 and PTPRD), and postsynaptic scaffolding (DLGAP1 and DLGAP2), which may be relevant to the pathogenesis of OCD. Four cases had CNVs involving known genomic disorder loci (1q21.1-21.2, 15q11.2-q13.1, 16p13.11, and 17p12). Further, we identified BTBD9 as a candidate gene for OCD. We also sequenced exomes of ten "CNV positive" trios and identified in one an additional plausibly relevant mutation: a 13 bp exonic deletion in DRD4. CONCLUSIONS Our findings suggest that rare CNVs may contribute to the etiology of OCD.
Collapse
Affiliation(s)
- Matthew J. Gazzellone
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - Mehdi Zarrei
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - Christie L. Burton
- Department of Psychiatry and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - Susan Walker
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - Mohammed Uddin
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - S. M. Shaheen
- Department of Psychiatry and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
- Mathison Centre for Mental Health Research and Education and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB Canada
| | - Julie Coste
- Department of Psychiatry and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - Rageen Rajendram
- Department of Psychiatry and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
- Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Reva J. Schachter
- Department of Psychiatry and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - Marlena Colasanto
- Department of Psychiatry and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - Gregory L. Hanna
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI USA
| | - David R. Rosenberg
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University, Detroit, MI USA
- The Children’s Hospital of Michigan, Detroit, MI USA
| | - Noam Soreni
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, St. Joseph’s Healthcare, Hamilton, ON Canada
| | - Kate D. Fitzgerald
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI USA
| | - Christian R. Marshall
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - Janet A. Buchanan
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - Daniele Merico
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| | - Paul D. Arnold
- Department of Psychiatry and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
- Mathison Centre for Mental Health Research and Education and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB Canada
- Departments of Psychiatry and Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB Canada
| | - Stephen W. Scherer
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
- Department of Molecular Genetics and McLaughlin Centre, University of Toronto, Toronto, ON Canada
| |
Collapse
|
131
|
Dworschak GC, Crétolle C, Hilger A, Engels H, Korsch E, Reutter H, Ludwig M. Comprehensive review of the duplication 3q syndrome and report of a patient with Currarino syndrome and de novo duplication 3q26.32-q27.2. Clin Genet 2016; 91:661-671. [PMID: 27549440 DOI: 10.1111/cge.12848] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 12/19/2022]
Abstract
Partial duplications of the long arm of chromosome 3, dup(3q), are a rare but well-described condition, sharing features of Cornelia de Lange syndrome. Around two thirds of cases are derived from unbalanced translocations, whereas pure dup(3q) have rarely been reported. Here, we provide an extensive review of the literature on dup(3q). This search revealed several patients with caudal malformations and anomalies, suggesting that caudal malformations or anomalies represent an inherent phenotypic feature of dup(3q). In this context, we report a patient with a pure de novo duplication 3q26.32-q27.2. The patient had the clinical diagnosis of Currarino syndrome (CS) (characterized by the triad of sacral anomalies, anorectal malformations and a presacral mass) and additional features, frequently detected in patients with a dup(3q). Mutations within the MNX1 gene were found to be causative in CS but no MNX1 mutation could be detected in our patient. Our comprehensive search for candidate genes located in the critical region of the duplication 3q syndrome, 3q26.3-q27, revealed a so far neglected phenotypic overlap of dup(3q) and the Pierpont syndrome, associated with a mutation of the TBL1XR1 gene on 3q26.32.
Collapse
Affiliation(s)
- G C Dworschak
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Pediatrics, Children's Hospital, University of Bonn, Bonn, Germany
| | - C Crétolle
- Department of Pediatric Surgery, Paris Descartes University, Paris, France.,National Reference Centre for Rare Diseases on Anorectal Malformations and Rare Pelvic Anomalies, Necker-Enfants Malades Hospital, Paris Descartes University, Paris, France
| | - A Hilger
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - H Engels
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - E Korsch
- Clinic for Pediatric Diseases, Kliniken der Stadt Köln GmbH, Cologne, Germany
| | - H Reutter
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
| | - M Ludwig
- Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| |
Collapse
|
132
|
Liu Y, Wang Z, De La Torre R, Barling A, Tsujikawa T, Hornick N, Hanifin J, Simpson E, Wang Y, Swanzey E, Wortham A, Ding H, Coussens LM, Kulesz-Martin M. Trim32 Deficiency Enhances Th2 Immunity and Predisposes to Features of Atopic Dermatitis. J Invest Dermatol 2016; 137:359-366. [PMID: 27720760 DOI: 10.1016/j.jid.2016.09.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 09/12/2016] [Accepted: 09/22/2016] [Indexed: 12/18/2022]
Abstract
Altered innate immunity is a feature of certain skin inflammatory diseases such as psoriasis and atopic dermatitis (AD). In this study, we provide evidence that deficiency in Trim32 (a tripartite motif [TRIM] protein with innate antiviral activity) contributes to a T helper type 2 biased response and predisposes to features of AD in mice. On treatment with the toll-like receptor 7 agonist imquimod (IMQ), Trim32 knockout mice displayed compromised psoriasiform phenotypes and defective T helper type 17 response. Instead, IMQ treatment of Trim32 knockout mice induced AD-like phenotypes with enhanced skin infiltration of eosinophils and mast cells, elevation of T helper type 2 cytokines/chemokines expression, and reduced expression of filaggrin protein expression. Furthermore, although the induction of phosphorylated Stat3 and RelA was compromised after IMQ treatment in the knockout mice, phosphorylated Stat6 was elevated. CC chemokine ligand 20 induction by tumor necrosis factor-α and IL-17A was reduced in Trim32-deficient keratinocytes, whereas CC chemokine ligand 5 induction by tumor necrosis factor-α and IL-4 was enhanced. In addition, Trim32 protein levels were elevated in mice treated with IMQ. Unlike Trim32 overexpression in psoriasis, TRIM32 levels were low in patients with AD. Based on Trim32 induction by IMQ, the lower levels of TRIM32 in AD skin compared with healthy control and psoriatic skin suggest a defective TRIM32 pathway in AD pathogenesis.
Collapse
Affiliation(s)
- Yuangang Liu
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA.
| | - Zhiping Wang
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Rachel De La Torre
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Ashley Barling
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Takahiro Tsujikawa
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon, USA
| | - Noah Hornick
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Jon Hanifin
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Eric Simpson
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Yun Wang
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Emily Swanzey
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Aaron Wortham
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Hao Ding
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lisa M Coussens
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon, USA
| | - Molly Kulesz-Martin
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA; Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon, USA.
| |
Collapse
|
133
|
Abstract
OBJECTIVE Children with ADHD have an increased risk of later developing personality disorders and criminal behavior. The object of the present review is to analyze the associations between ADHD and antisocial personality disorder (ASPD). METHOD A review of literature was done using EMBASE, PsycINFO, and Medline databases. RESULTS Eighteen prospective studies (n = 5,501) showed that ADHD with and without comorbid conduct disorder (CD) is a strong predictor for the risk of later development of antisocial personality disorder (ASPD). Some of the 13 cross-sectional/retrospective studies (n = 2,451) suggested that ADHD and CD might be a separate subtype of ADHD, that especially impulsivity in ADHD is a predictor for later development of ASPD, or that callous-unemotional traits in the ADHD children are called for a risk factor for later ASPD. CONCLUSION There is an increased risk for children with ADHD with or without comorbid CD to develop later onset of antisocial personality disorder.
Collapse
Affiliation(s)
- Ole Jakob Storebø
- Psychiatric Research Unit, Region Zealand, Holbæk, Denmark Child and Adolescent Psychiatric Department, Region Zealand, Holbæk, Denmark
| | - Erik Simonsen
- Child and Adolescent Psychiatric Department, Region Zealand, Holbæk, Denmark
| |
Collapse
|
134
|
Mak CCY, Chow PC, Liu APY, Chan KYK, Chu YWY, Mok GTK, Leung GKC, Yeung KS, Chau AKT, Lowther C, Scherer SW, Marshall CR, Bassett AS, Chung BHY. De novo large rare copy-number variations contribute to conotruncal heart disease in Chinese patients. NPJ Genom Med 2016; 1:16033. [PMID: 29263819 PMCID: PMC5685312 DOI: 10.1038/npjgenmed.2016.33] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/02/2016] [Accepted: 08/05/2016] [Indexed: 01/05/2023] Open
Abstract
Conotruncal heart anomalies (CTDs) are particularly prevalent congenital heart diseases (CHD) in Hong Kong. We surveyed large (>500 kb), rare (<1% frequency in controls) copy-number variations (CNVs) in Chinese patients with CTDs to identify potentially disease-causing variations. Adults who tested negative for 22q11.2 deletions were recruited from the adult CHD clinic in Hong Kong. Using a stringent calling criteria, high-confidence CNV calls were obtained, and a large control set comprising 3,987 Caucasian and 1,945 Singapore Chinese subjects was used to identify rare CNVs. Ten large rare CNVs were identified, and 3 in 108 individuals were confirmed to harbour de novo CNVs. All three patients were syndromic with a more complex phenotype, and each of these CNVs overlapped regions likely to be important in CHD. One was a 611 kb deletion at 17p13.3, telomeric to the Miller-Dieker syndrome (MDS) critical region, overlapping the NXN gene. Another was a 5 Mb deletion at 13q33.3, within a previously described critical region for CHD. A third CNV, previously unreported, was a large duplication at 2q22.3 overlapping the ZEB2 gene. The commonly reported 1q21.1 recurrent duplication was not observed in this Chinese cohort. We provide detailed phenotypic and genotypic descriptions of large rare genic CNVs that may represent CHD loci in the East Asian population. Larger samples of Chinese origin will be required to determine whether the genome-wide distribution differs from that found in predominantly European CHD cohorts.
Collapse
Affiliation(s)
- Christopher C Y Mak
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Pak Cheong Chow
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anthony P Y Liu
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kelvin Y K Chan
- Department of Obstetrics and Gynecology, Queen Mary Hospital, Hong Kong, China
| | - Yoyo W Y Chu
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Gary T K Mok
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Gordon K C Leung
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kit San Yeung
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Adolphus K T Chau
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chelsea Lowther
- The Clinical Genetics Research Program at The Centre for Addiction and Mental Health, The Dalglish Family 22q Clinic at The University Health Network, and The Department of Psychiatry at The University of Toronto, Toronto, ON, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christian R Marshall
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Anne S Bassett
- The Clinical Genetics Research Program at The Centre for Addiction and Mental Health, The Dalglish Family 22q Clinic at The University Health Network, and The Department of Psychiatry at The University of Toronto, Toronto, ON, Canada
| | - Brian H Y Chung
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
135
|
Tovo-Rodrigues L, Recamonde-Mendoza M, Paixão-Côrtes VR, Bruxel EM, Schuch JB, Friedrich DC, Rohde LA, Hutz MH. The role of protein intrinsic disorder in major psychiatric disorders. Am J Med Genet B Neuropsychiatr Genet 2016; 171:848-60. [PMID: 27184105 DOI: 10.1002/ajmg.b.32455] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/22/2016] [Indexed: 01/26/2023]
Abstract
Although new candidate genes for Autism Spectrum Disorder (ASD), Schizophrenia (SCZ), Attention-Deficit/Hyperactivity Disorder (ADHD), and Bipolar Disorder (BD) emerged from genome-wide association studies (GWAS), their underlying molecular mechanisms remain poorly understood. Evidences of the involvement of intrinsically disordered proteins in diseases have grown in the last decade. These proteins lack tridimensional structure under physiological conditions and are involved in important cellular functions such as signaling, recognition and regulation. The aim of the present study was to identify the role and abundance of intrinsically disordered proteins in a set of psychiatric diseases and to test whether diseases are different regarding protein intrinsic disorder. Our hypothesis is that differences across psychiatric illnesses phenotypes and symptoms may arise from differences in intrinsic protein disorder content and properties of each group. A bioinformatics prediction of intrinsic disorder was performed in proteins retrieved based on top findings from GWAS, Copy Number Variation and candidate gene investigations for each disease. This approach revealed that about 80% of studied proteins presented long stretches of disorder. This amount was significantly higher than that observed in general eukaryotic proteins, and those involved in cardiovascular diseases. These results suggest that proteins with intrinsic disorder are a common feature of neurodevelopment and synaptic transmission processes which are potentially involved in the etiology of psychiatric diseases. Moreover, we identified differences between ADHD and ASD when the binary prediction of structure and putative binding sites were compared. These differences may be related to variation in symptom complexity between both diseases. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Luciana Tovo-Rodrigues
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.,Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Mariana Recamonde-Mendoza
- Experimental and Molecular Cardiovascular Laboratory, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Estela M Bruxel
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Jaqueline B Schuch
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Deise C Friedrich
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luis A Rohde
- Child and Adolescent Psychiatric Division, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.,National Institute of Developmental Psychiatry for Children and Adolescents (INCT-CNPq), Brazil
| | - Mara H Hutz
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
136
|
Psychiatric gene discoveries shape evidence on ADHD's biology. Mol Psychiatry 2016; 21:1202-7. [PMID: 26573769 PMCID: PMC4820035 DOI: 10.1038/mp.2015.163] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/08/2015] [Accepted: 09/08/2015] [Indexed: 12/22/2022]
Abstract
A strong motivation for undertaking psychiatric gene discovery studies is to provide novel insights into unknown biology. Although attention-deficit hyperactivity disorder (ADHD) is highly heritable, and large, rare copy number variants (CNVs) contribute to risk, little is known about its pathogenesis and it remains commonly misunderstood. We assembled and pooled five ADHD and control CNV data sets from the United Kingdom, Ireland, United States of America, Northern Europe and Canada. Our aim was to test for enrichment of neurodevelopmental gene sets, implicated by recent exome-sequencing studies of (a) schizophrenia and (b) autism as a means of testing the hypothesis that common pathogenic mechanisms underlie ADHD and these other neurodevelopmental disorders. We also undertook hypothesis-free testing of all biological pathways. We observed significant enrichment of individual genes previously found to harbour schizophrenia de novo non-synonymous single-nucleotide variants (SNVs; P=5.4 × 10(-4)) and targets of the Fragile X mental retardation protein (P=0.0018). No enrichment was observed for activity-regulated cytoskeleton-associated protein (P=0.23) or N-methyl-D-aspartate receptor (P=0.74) post-synaptic signalling gene sets previously implicated in schizophrenia. Enrichment of ADHD CNV hits for genes impacted by autism de novo SNVs (P=0.019 for non-synonymous SNV genes) did not survive Bonferroni correction. Hypothesis-free testing yielded several highly significantly enriched biological pathways, including ion channel pathways. Enrichment findings were robust to multiple testing corrections and to sensitivity analyses that excluded the most significant sample. The findings reveal that CNVs in ADHD converge on biologically meaningful gene clusters, including ones now established as conferring risk of other neurodevelopmental disorders.
Collapse
|
137
|
Ramírez-Franco JJ, Munoz-Cuevas FJ, Luján R, Jurado S. Excitatory and Inhibitory Neurons in the Hippocampus Exhibit Molecularly Distinct Large Dense Core Vesicles. Front Cell Neurosci 2016; 10:202. [PMID: 27630542 PMCID: PMC5005380 DOI: 10.3389/fncel.2016.00202] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 08/05/2016] [Indexed: 12/21/2022] Open
Abstract
Hippocampal interneurons comprise a diverse family of inhibitory neurons that are critical for detailed information processing. Along with gamma-aminobutyric acid (GABA), interneurons secrete a myriad of neuroactive substances via secretory vesicles but the molecular composition and regulatory mechanisms remain largely unknown. In this study, we have carried out an immunohistofluorescence analysis to describe the molecular content of vesicles in distinct populations of hippocampal neurons. Our results indicate that phogrin, an integral protein of secretory vesicles in neuroendocrine cells, is highly enriched in parvalbumin-positive interneurons. Consistently, immunoelectron microscopy revealed phogrin staining in axon terminals of symmetrical synapses establishing inhibitory contacts with cell bodies of CA1 pyramidal neurons. Furthermore, phogrin is highly expressed in CA3 and dentate gyrus (DG) interneurons which are both positive for PV and neuropeptide Y. Surprisingly, chromogranin B a canonical large dense core vesicle marker, is excluded from inhibitory cells in the hippocampus but highly expressed in excitatory CA3 pyramidal neurons and DG granule cells. Our results provide the first evidence of phogrin expression in hippocampal interneurons and suggest the existence of molecularly distinct populations of secretory vesicles in different types of inhibitory neurons.
Collapse
Affiliation(s)
| | | | - Rafael Luján
- Synaptic Structure Laboratory, Departamento de Ciencias Médicas, Facultad de Medicina, Instituto de Investigación en Discapacidades Neurológicas, Universidad Castilla-La ManchaAlbacete, Spain
| | - Sandra Jurado
- Department of Pharmacology, University of Maryland School of MedicineBaltimore, MD, USA
| |
Collapse
|
138
|
Barnard-Brak L, Davis TN, Schmidt M, Richman DM. Effects associated with on- and off-label stimulant treatment of core autism and ADHD symptoms exhibited by children with autism spectrum disorder. Dev Neurorehabil 2016; 19:46-53. [PMID: 24739141 DOI: 10.3109/17518423.2014.904949] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Families of children with autism spectrum disorder are barraged by different treatment options. Some of these options have the support of empirical evidence while others do not. Stimulant treatments are typically utilized to treat symptoms of ADHD indicating an on-label use of such treatment. METHODS This study examines the association of stimulant treatment with the on- (symptoms of ADHD) and off- (symptoms of ASD) label symptoms among children with ASD via a non-clinical, population-based sample. RESULTS Results indicate no significant association of stimulant treatment with a reduction of on- or off-label symptoms among children with ASD. CONCLUSION Stimulant medications utilized in the treatment of DSM core symptoms of autism spectrum disorder would be considered an off-label use because there is limited evidence to support that stimulants are effective in treating core symptoms of ASD, which is supported by the results of the current study.
Collapse
Affiliation(s)
- Lucy Barnard-Brak
- a College of Education, Texas Tech University , Lubbock , Texas , USA
| | - Tonya N Davis
- b Department of Educational Psychology , School of Education, Baylor University , Waco , Texas , USA , and
| | | | | |
Collapse
|
139
|
Mosca SJ, Langevin LM, Dewey D, Innes AM, Lionel AC, Marshall CC, Scherer SW, Parboosingh JS, Bernier FP. Copy-number variations are enriched for neurodevelopmental genes in children with developmental coordination disorder. J Med Genet 2016; 53:812-819. [DOI: 10.1136/jmedgenet-2016-103818] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 01/24/2023]
|
140
|
Torrico B, Chiocchetti AG, Bacchelli E, Trabetti E, Hervás A, Franke B, Buitelaar JK, Rommelse N, Yousaf A, Duketis E, Freitag CM, Caballero-Andaluz R, Martinez-Mir A, Scholl FG, Ribasés M, Battaglia A, Malerba G, Delorme R, Benabou M, Maestrini E, Bourgeron T, Cormand B, Toma C. Lack of replication of previous autism spectrum disorder GWAS hits in European populations. Autism Res 2016; 10:202-211. [DOI: 10.1002/aur.1662] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/15/2016] [Accepted: 06/03/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Bàrbara Torrico
- Departament de Genètica; Microbiologia i Estadística, Universitat de Barcelona; Av. Diagonal 643 08028 Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III; C/ Monforte de Lemos 3-5 28029 Madrid Spain
- Institut de Biomedicina, Universitat de Barcelona; Av. Diagonal 643 08028 Barcelona Spain
- Institut de Recerca Pediàtrica Hospital Sant Joan de Dáu; Santa Rosa 39-57 08950 Esplugues de Llobregat Spain
| | - Andreas G. Chiocchetti
- Department of Child and Adolescent Psychiatry; Psychosomatics and Psychotherapy, Autism Research and Intervention Center of Excellence Frankfurt, JW Goethe University; Deutschordenstraße 50 60528 Frankfurt am Main Frankfurt am Main Germany
| | - Elena Bacchelli
- Department of Pharmacy and Biotechnology; University of Bologna; via Selmi 3 40126 Bologna Italy
| | - Elisabetta Trabetti
- Department of Neurological; Biomedical and Movement Sciences, Section of Biology and Genetics, University of Verona; Strada le Grazie 8 37134 Verona Italy
| | - Amaia Hervás
- Child and Adolescent Mental Health Unit; University Hospital MutuaTerrassa; Plaza del Dr Robert s/n 08221 Terrassa Barcelona Spain
| | - Barbara Franke
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Human Genetics; Geert Grooteplein-Zuid 10 6525 GA Nijmegen The Netherlands
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Psychiatry; Geert Grooteplein-Zuid 10 6525 GA Nijmegen The Netherlands
| | - Jan K. Buitelaar
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience; Geert Grooteplein Noord 21 6525 EZ Nijmegen The Netherlands
- Karakter Child and Adolescent Psychiatry University Center; Reinier Postlaan 12 6525 GC Nijmegen The Netherlands
| | - Nanda Rommelse
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Psychiatry; Geert Grooteplein-Zuid 10 6525 GA Nijmegen The Netherlands
- Karakter Child and Adolescent Psychiatry University Center; Reinier Postlaan 12 6525 GC Nijmegen The Netherlands
| | - Afsheen Yousaf
- Department of Child and Adolescent Psychiatry; Psychosomatics and Psychotherapy, Autism Research and Intervention Center of Excellence Frankfurt, JW Goethe University; Deutschordenstraße 50 60528 Frankfurt am Main Frankfurt am Main Germany
- Institute for Molecular Bioinformatics; Johann Wolfgang Goethe-University; Robert-Mayer-Str. 11-15 60325 Frankfurt am Main Germany
| | - Eftichia Duketis
- Department of Child and Adolescent Psychiatry; Psychosomatics and Psychotherapy, Autism Research and Intervention Center of Excellence Frankfurt, JW Goethe University; Deutschordenstraße 50 60528 Frankfurt am Main Frankfurt am Main Germany
| | - Christine M. Freitag
- Department of Child and Adolescent Psychiatry; Psychosomatics and Psychotherapy, Autism Research and Intervention Center of Excellence Frankfurt, JW Goethe University; Deutschordenstraße 50 60528 Frankfurt am Main Frankfurt am Main Germany
| | | | - Amalia Martinez-Mir
- Instituto de Biomedicina de Sevilla (IBiS); Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla; Av. Manuel Siurot s/n 41013 Seville Spain
| | - Francisco G. Scholl
- Instituto de Biomedicina de Sevilla (IBiS); Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla; Av. Manuel Siurot s/n 41013 Seville Spain
| | - Marta Ribasés
- Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona; Passeig Vall d'Hebron 119-129 08035 Barcelona Spain
- Department of Psychiatry; Hospital Universitari Vall d'Hebron; Passeig Vall d'Hebron 119-129, 08035 Barcelona Spain
- Biomedical Network Research Center on Mental Health (CIBERSAM); Av. Monforte de Lemos, 3-5 28029 Madrid Spain
| | - Agatino Battaglia
- Stella Maris Clinical Research Institute for Child and Adolescent Neuropsychiatry, via dei Giacinti 2, 56128 Calambrone; Pisa Italy
| | - Giovanni Malerba
- Department of Neurological; Biomedical and Movement Sciences, Section of Biology and Genetics, University of Verona; Strada le Grazie 8 37134 Verona Italy
| | - Richard Delorme
- Institut Pasteur, Human Genetics and Cognitive Functions Unit; 25, rue du docteur Roux 75015 Paris France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur; 25, rue du docteur Roux 75015 Paris France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions; 5 Rue Thomas Mann 75013 Paris France
- Assistance Publique-Hôpitaux de Paris, Child and Adolescent Psychiatry Department, Robert Debré Hospital; 48Bd Sérurier 75019 Paris France
| | - Marion Benabou
- Institut Pasteur, Human Genetics and Cognitive Functions Unit; 25, rue du docteur Roux 75015 Paris France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur; 25, rue du docteur Roux 75015 Paris France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions; 5 Rue Thomas Mann 75013 Paris France
| | - Elena Maestrini
- Department of Pharmacy and Biotechnology; University of Bologna; via Selmi 3 40126 Bologna Italy
| | - Thomas Bourgeron
- Institut Pasteur, Human Genetics and Cognitive Functions Unit; 25, rue du docteur Roux 75015 Paris France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur; 25, rue du docteur Roux 75015 Paris France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions; 5 Rue Thomas Mann 75013 Paris France
| | - Bru Cormand
- Departament de Genètica; Microbiologia i Estadística, Universitat de Barcelona; Av. Diagonal 643 08028 Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III; C/ Monforte de Lemos 3-5 28029 Madrid Spain
- Institut de Biomedicina, Universitat de Barcelona; Av. Diagonal 643 08028 Barcelona Spain
- Institut de Recerca Pediàtrica Hospital Sant Joan de Dáu; Santa Rosa 39-57 08950 Esplugues de Llobregat Spain
| | - Claudio Toma
- Departament de Genètica; Microbiologia i Estadística, Universitat de Barcelona; Av. Diagonal 643 08028 Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III; C/ Monforte de Lemos 3-5 28029 Madrid Spain
- Institut de Biomedicina, Universitat de Barcelona; Av. Diagonal 643 08028 Barcelona Spain
- Neuroscience Research Australia; Barker St Randwick 2031 Sydney New South Wales Australia
- School of Medical Sciences, University of New South Wales; High St, Kensington 2052 Sydney New South Wales Australia
| | | |
Collapse
|
141
|
van der Plas E, Dupuis A, Arnold P, Crosbie J, Schachar R. Association of Autism Spectrum Disorder with Obsessive-Compulsive and Attention-Deficit/Hyperactivity Traits and Response Inhibition in a Community Sample. J Autism Dev Disord 2016; 46:3115-25. [DOI: 10.1007/s10803-016-2853-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
142
|
Abstract
PURPOSE OF REVIEW The following review provides some description of the movement in cross-disorder psychiatric genomics toward addressing both comorbidity and polygenicity. RECENT FINDINGS We attempt to show how dimensional approaches to the phenotype have led to further addressing the problem of comorbidity of psychiatric diagnoses. And we also attempt to show how a dimensional approach to the genome, with different statistical methods from traditional genome-wide association analyses, has begun to resolve the problem of massive polygenicity. SUMMARY Cross-disorder research, of any area in psychiatry, arguably has the most potential to inform clinical diagnosis, early detection and prevention strategies, and pharmacological treatment research. Future research might leverage what we now know to inform developmental studies of risk and resilience.
Collapse
Affiliation(s)
- Anna R Docherty
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Arden A Moscati
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Ayman H Fanous
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Washington Veterans Affairs Healthcare System, Washington D.C., USA; Georgetown University School of Medicine, Washington D.C., USA
| |
Collapse
|
143
|
Gallo EF, Posner J. Moving towards causality in attention-deficit hyperactivity disorder: overview of neural and genetic mechanisms. Lancet Psychiatry 2016; 3:555-67. [PMID: 27183902 PMCID: PMC4893880 DOI: 10.1016/s2215-0366(16)00096-1] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/04/2016] [Accepted: 03/04/2016] [Indexed: 12/17/2022]
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder characterised by developmentally inappropriate levels of inattention and hyperactivity or impulsivity. The heterogeneity of its clinical manifestations and the differential responses to treatment and varied prognoses have long suggested myriad underlying causes. Over the past decade, clinical and basic research efforts have uncovered many behavioural and neurobiological alterations associated with ADHD, from genes to higher order neural networks. Here, we review the neurobiology of ADHD by focusing on neural circuits implicated in the disorder and discuss how abnormalities in circuitry relate to symptom presentation and treatment. We summarise the literature on genetic variants that are potentially related to the development of ADHD, and how these, in turn, might affect circuit function and relevant behaviours. Whether these underlying neurobiological factors are causally related to symptom presentation remains unresolved. Therefore, we assess efforts aimed at disentangling issues of causality, and showcase the shifting research landscape towards endophenotype refinement in clinical and preclinical settings. Furthermore, we review approaches being developed to understand the neurobiological underpinnings of this complex disorder, including the use of animal models, neuromodulation, and pharmacoimaging studies.
Collapse
Affiliation(s)
- Eduardo F Gallo
- Columbia University and New York State Psychiatric Institute, New York, NY, USA.
| | - Jonathan Posner
- Columbia University and New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
144
|
Rosenfeld JA, Patel A. Chromosomal Microarrays: Understanding Genetics of Neurodevelopmental Disorders and Congenital Anomalies. J Pediatr Genet 2016; 6:42-50. [PMID: 28180026 DOI: 10.1055/s-0036-1584306] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/23/2016] [Indexed: 01/09/2023]
Abstract
Chromosomal microarray (CMA) testing, used to identify DNA copy number variations (CNVs), has helped advance knowledge about genetics of human neurodevelopmental disease and congenital anomalies. It has aided in discovering new CNV syndromes and uncovering disease genes. It has discovered CNVs that are not fully penetrant and/or cause a spectrum of phenotypes, including intellectual disability, autism, schizophrenia, and dysmorphisms. Such CNVs can pose challenges to genetic counseling. They also have helped increase knowledge of genetic risk factors for neurodevelopmental disease and raised awareness of possible shared etiologies among these variable phenotypes. Advances in CMA technology allow CNV identification at increasingly finer scales, improving detection of pathogenic changes, although these sometimes are difficult to distinguish from normal population variation. This paper confronts some of the challenges uncovered by CMA testing while reviewing advances in genetics and the clinical use of this test that has replaced standard karyotyping in most genetic evaluations.
Collapse
Affiliation(s)
- Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States; Baylor Miraca Genetics Laboratories, Baylor College of Medicine, Houston, Texas, United States
| | - Ankita Patel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States; Baylor Miraca Genetics Laboratories, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
145
|
Lowther C, Speevak M, Armour CM, Goh ES, Graham GE, Li C, Zeesman S, Nowaczyk MJM, Schultz LA, Morra A, Nicolson R, Bikangaga P, Samdup D, Zaazou M, Boyd K, Jung JH, Siu V, Rajguru M, Goobie S, Tarnopolsky MA, Prasad C, Dick PT, Hussain AS, Walinga M, Reijenga RG, Gazzellone M, Lionel AC, Marshall CR, Scherer SW, Stavropoulos DJ, McCready E, Bassett AS. Molecular characterization of NRXN1 deletions from 19,263 clinical microarray cases identifies exons important for neurodevelopmental disease expression. Genet Med 2016; 19:53-61. [PMID: 27195815 PMCID: PMC4980119 DOI: 10.1038/gim.2016.54] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/16/2016] [Indexed: 01/31/2023] Open
Abstract
Purpose The purpose of the current study was to assess the penetrance of NRXN1 deletions. Methods We compared the prevalence and genomic extent of NRXN1 deletions identified among 19,263 clinically referred cases to that of 15,264 controls. The burden of additional clinically relevant CNVs was used as a proxy to estimate the relative penetrance of NRXN1 deletions. Results We identified 41 (0.21%) previously unreported exonic NRXN1 deletions ascertained for developmental delay/intellectual disability, significantly greater than in controls [OR=8.14 (95% CI 2.91–22.72), p< 0.0001)]. Ten (22.7%) of these had a second clinically relevant CNV. Subjects with a deletion near the 3′ end of NRXN1 were significantly more likely to have a second rare CNV than subjects with a 5′ NRXN1 deletion [OR=7.47 (95% CI 2.36–23.61), p=0.0006]. The prevalence of intronic NRXN1 deletions was not statistically different between cases and controls (p=0.618). The majority (63.2%) of intronic NRXN1 deletion cases had a second rare CNV, a two-fold greater prevalence than for exonic NRXN1 deletion cases (p=0.0035). Conclusions The results support the importance of exons near the 5′ end of NRXN1 in the expression of neurodevelopmental disorders. Intronic NRXN1 deletions do not appear to substantially increase the risk for clinical phenotypes.
Collapse
Affiliation(s)
- Chelsea Lowther
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Marsha Speevak
- Trillium Health Partners Credit Valley Site, Toronto, Ontario, Canada
| | - Christine M Armour
- Regional Genetics Program, Children's Hospital of Eastern Ontario, Toronto, ON, Canada
| | - Elaine S Goh
- Trillium Health Partners Credit Valley Site, Toronto, Ontario, Canada
| | - Gail E Graham
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Chumei Li
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada.,McMaster Children's Hospital, Department of Pediatrics and Clinical Genetics Program, Hamilton, Ontario, Canada
| | - Susan Zeesman
- McMaster Children's Hospital, Department of Pediatrics and Clinical Genetics Program, Hamilton, Ontario, Canada
| | - Malgorzata J M Nowaczyk
- McMaster Children's Hospital, Department of Pediatrics and Clinical Genetics Program, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Lee-Anne Schultz
- McMaster Children's Hospital, Department of Pediatrics and Clinical Genetics Program, Hamilton, Ontario, Canada
| | - Antonella Morra
- Trillium Health Partners Credit Valley Site, Toronto, Ontario, Canada
| | - Rob Nicolson
- Department of Psychiatry, Western University, London, Ontario, Canada
| | | | - Dawa Samdup
- Hotel Dieu Hospital, Child Development Centre, Kingston, Ontario, Canada
| | - Mostafa Zaazou
- Trillium Health Partners Credit Valley Site, Toronto, Ontario, Canada
| | - Kerry Boyd
- Department of Psychiatry, McMaster University, Hamilton, Ontario, Canada
| | - Jack H Jung
- London Health Sciences Centre, Children's Hospital of Western Ontario, London, Ontario, Canada
| | - Victoria Siu
- Department of Pediatrics, Schulich School of Medicine and Dentistry, London, Ontario, Canada
| | | | - Sharan Goobie
- Department of Pediatrics, Schulich School of Medicine and Dentistry, London, Ontario, Canada
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Chitra Prasad
- Department of Pediatrics, Schulich School of Medicine and Dentistry, London, Ontario, Canada
| | - Paul T Dick
- Grey Bruce Health Services, Owen Sound, Ontario, Canada
| | - Asmaa S Hussain
- London Health Sciences Centre, Children's Hospital of Western Ontario, London, Ontario, Canada
| | | | | | - Matthew Gazzellone
- The Centre for Applied Genomics, the Hospital for Sick Children, Toronto, Ontario, Canada
| | - Anath C Lionel
- The Centre for Applied Genomics, the Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christian R Marshall
- The Centre for Applied Genomics, the Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics, the Hospital for Sick Children, Toronto, Ontario, Canada.,McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Dimitri J Stavropoulos
- Cytogenetics Laboratory, Department of Pediatric Laboratory Medicine, the Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elizabeth McCready
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Anne S Bassett
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
146
|
D'Arrigo S, Gavazzi F, Alfei E, Zuffardi O, Montomoli C, Corso B, Buzzi E, Sciacca FL, Bulgheroni S, Riva D, Pantaleoni C. The Diagnostic Yield of Array Comparative Genomic Hybridization Is High Regardless of Severity of Intellectual Disability/Developmental Delay in Children. J Child Neurol 2016; 31:691-9. [PMID: 26511719 DOI: 10.1177/0883073815613562] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/22/2015] [Indexed: 12/08/2022]
Abstract
Microarray-based comparative genomic hybridization is a method of molecular analysis that identifies chromosomal anomalies (or copy number variants) that correlate with clinical phenotypes. The aim of the present study was to apply a clinical score previously designated by de Vries to 329 patients with intellectual disability/developmental disorder (intellectual disability/developmental delay) referred to our tertiary center and to see whether the clinical factors are associated with a positive outcome of aCGH analyses. Another goal was to test the association between a positive microarray-based comparative genomic hybridization result and the severity of intellectual disability/developmental delay. Microarray-based comparative genomic hybridization identified structural chromosomal alterations responsible for the intellectual disability/developmental delay phenotype in 16% of our sample. Our study showed that causative copy number variants are frequently found even in cases of mild intellectual disability (30.77%). We want to emphasize the need to conduct microarray-based comparative genomic hybridization on all individuals with intellectual disability/developmental delay, regardless of the severity, because the degree of intellectual disability/developmental delay does not predict the diagnostic yield of microarray-based comparative genomic hybridization.
Collapse
Affiliation(s)
- Stefano D'Arrigo
- Developmental Neurology Department, IRCCS Fondazione Istituto Neurologico "C. Besta," Milan, Italy
| | - Francesco Gavazzi
- Developmental Neurology Department, IRCCS Fondazione Istituto Neurologico "C. Besta," Milan, Italy
| | - Enrico Alfei
- Developmental Neurology Department, IRCCS Fondazione Istituto Neurologico "C. Besta," Milan, Italy
| | | | - Cristina Montomoli
- Department of Public Health, Neuroscience, Experimental and Forensic Medicine, University of Pavia, Italy
| | - Barbara Corso
- Neuroscience Institute, National Research Council, Padua, Italy
| | - Erika Buzzi
- Institute of Neurological and Psychiatric Sciences of Childhood and Adolescence, University of Milan, A.O. San Paolo, Milan, Italy
| | - Francesca L Sciacca
- Medical Genetics Department, IRCCS Fondazione Istituto Neurologico "C. Besta," Milan, Italy
| | - Sara Bulgheroni
- Developmental Neurology Department, IRCCS Fondazione Istituto Neurologico "C. Besta," Milan, Italy
| | - Daria Riva
- Developmental Neurology Department, IRCCS Fondazione Istituto Neurologico "C. Besta," Milan, Italy
| | - Chiara Pantaleoni
- Developmental Neurology Department, IRCCS Fondazione Istituto Neurologico "C. Besta," Milan, Italy
| |
Collapse
|
147
|
Exploiting aberrant mRNA expression in autism for gene discovery and diagnosis. Hum Genet 2016; 135:797-811. [DOI: 10.1007/s00439-016-1673-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/17/2016] [Indexed: 01/09/2023]
|
148
|
Dewey D, Bernier FP. The Concept of Atypical Brain Development in Developmental Coordination Disorder (DCD)—a New Look. CURRENT DEVELOPMENTAL DISORDERS REPORTS 2016. [DOI: 10.1007/s40474-016-0086-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
149
|
Berkowicz SR, Featherby TJ, Qu Z, Giousoh A, Borg NA, Heng JI, Whisstock JC, Bird PI. Brinp1(-/-) mice exhibit autism-like behaviour, altered memory, hyperactivity and increased parvalbumin-positive cortical interneuron density. Mol Autism 2016; 7:22. [PMID: 27042284 PMCID: PMC4818446 DOI: 10.1186/s13229-016-0079-7] [Citation(s) in RCA: 30] [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: 12/02/2015] [Accepted: 02/11/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND BMP/RA-inducible neural-specific protein 1 (Brinp1) is highly conserved in vertebrates, and continuously expressed in the neocortex, hippocampus, olfactory bulb and cerebellum from mid-embryonic development through to adulthood. METHODS Brinp1 knock-out (Brinp1(-/-)) mice were generated by Cre-recombinase-mediated removal of the third exon of Brinp1. Knock-out mice were characterised by behavioural phenotyping, immunohistochemistry and expression analysis of the developing and adult brain. RESULTS Absence of Brinp1 during development results in a behavioural phenotype resembling autism spectrum disorder (ASD), in which knock-out mice show reduced sociability and changes in vocalisation capacity. In addition, Brinp1(-/-) mice exhibit hyper-locomotor activity, have impaired short-term memory, and exhibit poor reproductive success. Brinp1(-/-) mice show increased density of parvalbumin-expressing interneurons in the adult mouse brain. Brinp1(-/-) mice do not show signs of altered neural precursor proliferation or increased apoptosis during late embryonic brain development. The expression of the related neuronal migration genes Astn1 and Astn2 is increased in the brains of Brinp1(-/-) mice, suggesting that they may ameliorate the effects of Brinp1 loss. CONCLUSIONS Brinp1 plays an important role in normal brain development and function by influencing neuronal distribution within the cortex. The increased cortical PV-positive interneuron density and altered behaviour of Brinp1(-/-) mice resemble features of a subset of human neurological disorders; namely autism spectrum disorder (ASD) and the hyperactivity aspect of attention deficit hyperactivity disorder (ADHD).
Collapse
Affiliation(s)
- Susan R. Berkowicz
- />Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
| | - Travis J. Featherby
- />Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052 Australia
| | - Zhengdong Qu
- />Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800 Australia
| | - Aminah Giousoh
- />Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
| | - Natalie A. Borg
- />Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
| | - Julian I. Heng
- />Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800 Australia
| | - James C. Whisstock
- />Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
- />Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC 3800 Australia
| | - Phillip I. Bird
- />Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
| |
Collapse
|
150
|
Abstract
Attention deficit hyperactivity disorder (ADHD) is a childhood-onset neurodevelopmental disorder with a prevalence of 1·4-3·0%. It is more common in boys than girls. Comorbidity with childhood-onset neurodevelopmental disorders and psychiatric disorders is substantial. ADHD is highly heritable and multifactorial; multiple genes and non-inherited factors contribute to the disorder. Prenatal and perinatal factors have been implicated as risks, but definite causes remain unknown. Most guidelines recommend a stepwise approach to treatment, beginning with non-drug interventions and then moving to pharmacological treatment in those most severely affected. Randomised controlled trials show short-term benefits of stimulant medication and atomoxetine. Meta-analyses of blinded trials of non-drug treatments have not yet proven the efficacy of such interventions. Longitudinal studies of ADHD show heightened risk of multiple mental health and social difficulties as well as premature mortality in adult life.
Collapse
Affiliation(s)
- Anita Thapar
- Child & Adolescent Psychiatry Section, Institute of Psychological Medicine and Clinical Neurosciences, and MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cardiff, UK.
| | - Miriam Cooper
- Child & Adolescent Psychiatry Section, Institute of Psychological Medicine and Clinical Neurosciences, and MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|