1
|
Gomez L, Cadilhac C, Prados J, Mule N, Jabaudon D, Dayer A. Developmental emergence of cortical neurogliaform cell diversity. Development 2023; 150:dev201830. [PMID: 37401408 PMCID: PMC10445751 DOI: 10.1242/dev.201830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/17/2023] [Indexed: 07/05/2023]
Abstract
GABAergic interneurons are key regulators of cortical circuit function. Among the dozens of reported transcriptionally distinct subtypes of cortical interneurons, neurogliaform cells (NGCs) are unique: they are recruited by long-range excitatory inputs, are a source of slow cortical inhibition and are able to modulate the activity of large neuronal populations. Despite their functional relevance, the developmental emergence and diversity of NGCs remains unclear. Here, by combining single-cell transcriptomics, genetic fate mapping, and electrophysiological and morphological characterization, we reveal that discrete molecular subtypes of NGCs, with distinctive anatomical and molecular profiles, populate the mouse neocortex. Furthermore, we show that NGC subtypes emerge gradually through development, as incipient discriminant molecular signatures are apparent in preoptic area (POA)-born NGC precursors. By identifying NGC developmentally conserved transcriptional programs, we report that the transcription factor Tox2 constitutes an identity hallmark across NGC subtypes. Using CRISPR-Cas9-mediated genetic loss of function, we show that Tox2 is essential for NGC development: POA-born cells lacking Tox2 fail to differentiate into NGCs. Together, these results reveal that NGCs are born from a spatially restricted pool of Tox2+ POA precursors, after which intra-type diverging molecular programs are gradually acquired post-mitotically and result in functionally and molecularly discrete NGC cortical subtypes.
Collapse
Affiliation(s)
- Lucia Gomez
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva, Switzerland
| | - Christelle Cadilhac
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva, Switzerland
| | - Julien Prados
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva, Switzerland
| | - Nandkishor Mule
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva, Switzerland
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva, Switzerland
- Clinic of Neurology, Geneva University Hospital, 1211 Geneva, Switzerland
| | - Alexandre Dayer
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva, Switzerland
- Department of Psychiatry, Geneva University Hospital, 1205 Geneva, Switzerland
| |
Collapse
|
2
|
Wu BS, Ge YJ, Zhang W, Chen SD, Xiang ST, Zhang YR, Ou YN, Jiang YC, Tan L, Cheng W, Suckling J, Feng JF, Yu JT, Mao Y. Genome-wide association study of cerebellar white matter microstructure and genetic overlap with common brain disorders. Neuroimage 2023; 269:119928. [PMID: 36740028 DOI: 10.1016/j.neuroimage.2023.119928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/12/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The cerebellum is recognized as being involved in neurocognitive and motor functions with communication with extra-cerebellar regions relying on the white matter integrity of the cerebellar peduncles. However, the genetic determinants of cerebellar white matter integrity remain largely unknown. METHODS We conducted a genome-wide association analysis of cerebellar white matter microstructure using diffusion tensor imaging data from 25,415 individuals from UK Biobank. The integrity of cerebellar white matter microstructure was measured as fractional anisotropy (FA) and mean diffusivity (MD). Identification of independent genomic loci, functional annotation, and tissue and cell-type analysis were conducted with FUMA. The linkage disequilibrium score regression (LDSC) was used to calculate genetic correlations between cerebellar white matter microstructure and regional brain volumes and brain-related traits. Furthermore, the conditional/conjunctional false discovery rate (condFDR/conjFDR) framework was employed to identify the shared genetic basis between cerebellar white matter microstructure and common brain disorders. RESULTS We identified 11 genetic loci (P < 8.3 × 10-9) and 86 genes associated with cerebellar white matter microstructure. Further functional enrichment analysis implicated the involvement of GABAergic neurons and cholinergic pathways. Significant polygenetic overlap between cerebellar white matter tracts and their anatomically connected or adjacent brain regions was detected. In addition, we report the overall genetic correlation and specific loci shared between cerebellar white matter microstructural integrity and brain-related traits, including movement, cognitive, psychiatric, and cerebrovascular categories. CONCLUSIONS Collectively, this study represents a step forward in understanding the genetics of cerebellar white matter microstructure and its shared genetic etiology with common brain disorders.
Collapse
Affiliation(s)
- Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Jun Ge
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Tong Xiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu-Chao Jiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China; Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - John Suckling
- Department of Psychiatry, Brain Mapping Unit, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Ying Mao
- Department of Neurosurgery and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Odintsova VV, Hagenbeek FA, van der Laan CM, van de Weijer S, Boomsma DI. Genetics and epigenetics of human aggression. HANDBOOK OF CLINICAL NEUROLOGY 2023; 197:13-44. [PMID: 37633706 DOI: 10.1016/b978-0-12-821375-9.00005-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2023]
Abstract
There is substantial variation between humans in aggressive behavior, with its biological etiology and molecular genetic basis mostly unknown. This review chapter offers an overview of genomic and omics studies revealing the genetic contribution to aggression and first insights into associations with epigenetic and other omics (e.g., metabolomics) profiles. We allowed for a broad phenotype definition including studies on "aggression," "aggressive behavior," or "aggression-related traits," "antisocial behavior," "conduct disorder," and "oppositional defiant disorder." Heritability estimates based on family and twin studies in children and adults of this broadly defined phenotype of aggression are around 50%, with relatively small fluctuations around this estimate. Next, we review the genome-wide association studies (GWAS) which search for associations with alleles and also allow for gene-based tests and epigenome-wide association studies (EWAS) which seek to identify associations with differently methylated regions across the genome. Both GWAS and EWAS allow for construction of Polygenic and DNA methylation scores at an individual level. Currently, these predict a small percentage of variance in aggression. We expect that increases in sample size will lead to additional discoveries in GWAS and EWAS, and that multiomics approaches will lead to a more comprehensive understanding of the molecular underpinnings of aggression.
Collapse
Affiliation(s)
- Veronika V Odintsova
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, The Netherlands; Mental Health Division, Amsterdam Public Health (APH) Research Institute, Amsterdam, The Netherlands
| | - Fiona A Hagenbeek
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Mental Health Division, Amsterdam Public Health (APH) Research Institute, Amsterdam, The Netherlands
| | - Camiel M van der Laan
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Netherlands Institute for the Study of Crime and Law Enforcement (NSCR), Amsterdam, The Netherlands
| | - Steve van de Weijer
- Netherlands Institute for the Study of Crime and Law Enforcement (NSCR), Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Luo L, Cai Y, Zhang Y, Hsu CG, Korshunov VA, Long X, Knight PA, Berk BC, Yan C. Role of PDE10A in vascular smooth muscle cell hyperplasia and pathological vascular remodelling. Cardiovasc Res 2022; 118:2703-2717. [PMID: 34550322 PMCID: PMC9890476 DOI: 10.1093/cvr/cvab304] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 09/17/2021] [Indexed: 02/05/2023] Open
Abstract
AIMS Intimal hyperplasia is a common feature of vascular remodelling disorders. Accumulation of synthetic smooth muscle cell (SMC)-like cells is the main underlying cause. Current therapeutic approaches including drug-eluting stents are not perfect due to the toxicity on endothelial cells and novel therapeutic strategies are needed. Our preliminary screening for dysregulated cyclic nucleotide phosphodiesterases (PDEs) in growing SMCs revealed the alteration of PDE10A expression. Herein, we investigated the function of PDE10A in SMC proliferation and intimal hyperplasia both in vitro and in vivo. METHODS AND RESULTS RT-qPCR, immunoblot, and in situ proximity ligation assay were performed to determine PDE10A expression in synthetic SMCs and injured vessels. We found that PDE10A mRNA and/or protein levels are up-regulated in cultured SMCs upon growth stimulation, as well as in intimal cells in injured mouse femoral arteries. To determine the cellular functions of PDE10A, we focused on its role in SMC proliferation. The anti-mitogenic effects of PDE10A on SMCs were evaluated via cell counting, BrdU incorporation, and flow cytometry. We found that PDE10A deficiency or inhibition arrested the SMC cell cycle at G1-phase with a reduction of cyclin D1. The anti-mitotic effect of PDE10A inhibition was dependent on cGMP-dependent protein kinase Iα (PKGIα), involving C-natriuretic peptide (CNP) and particulate guanylate cyclase natriuretic peptide receptor 2 (NPR2). In addition, the effects of genetic depletion and pharmacological inhibition of PDE10A on neointimal formation were examined in a mouse model of femoral artery wire injury. Both PDE10A knockout and inhibition decreased injury-induced intimal thickening in femoral arteries by at least 50%. Moreover, PDE10A inhibition decreased ex vivo remodelling of cultured human saphenous vein segments. CONCLUSIONS Our findings indicate that PDE10A contributes to SMC proliferation and intimal hyperplasia at least partially via antagonizing CNP/NPR2/cGMP/PKG1α signalling and suggest that PDE10A may be a novel drug target for treating vascular occlusive disease.
Collapse
Affiliation(s)
- Lingfeng Luo
- Department of Biochemistry and Biophysics, University of Rochester School
of Medicine and Dentistry, Rochester, NY,
USA
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Yujun Cai
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Yishuai Zhang
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Chia G Hsu
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Vyacheslav A Korshunov
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Xiaochun Long
- Department of Vascular Biology Center and Medicine, Medical College of
Georgia, Augusta, GA, USA
| | - Peter A Knight
- Department of Surgery, University of Rochester School of Medicine and
Dentistry, Rochester, NY, USA
| | - Bradford C Berk
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Chen Yan
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| |
Collapse
|
5
|
Nedic Erjavec G, Tudor L, Nikolac Perkovic M, Podobnik J, Dodig Curkovic K, Curkovic M, Svob Strac D, Cusek M, Bortolato M, Pivac N. Serotonin 5-HT 2A receptor polymorphisms are associated with irritability and aggression in conduct disorder. Prog Neuropsychopharmacol Biol Psychiatry 2022; 117:110542. [PMID: 35257831 DOI: 10.1016/j.pnpbp.2022.110542] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/20/2022] [Accepted: 02/27/2022] [Indexed: 01/01/2023]
Abstract
In childhood and adolescence, overt antisocial and aggressive manifestations are typically diagnosed as conduct disorder (CD). Given that the emerging research has pointed to the influence of 5-HT2A receptors in the ontogeny of aggression, we aimed to analyze the association of its genetic polymorphisms with CD. The study included 228 male adolescent subjects (120 with and 108 without CD). CD was diagnosed according to Structured Clinical Interview for DSM-IV criteria, while evaluations of aggressive/dissociative behaviors were performed using psychometric questionnaires including the PCL-YV, OAS-M, KADS, and CBCL. Platelet 5-HT concentration was determined by spectrophotofluorometry. Genotyping of 5-HT2A receptor polymorphisms rs2070040, rs9534511, rs4142900, rs9534512 was performed using TaqMan SNP Genotyping Assays. Subjective irritability, physical aggression toward others, and antisocial behavior were strongly associated with the G allele of rs2070040 and rs4142900, and the C allele of rs9534511 and rs9534512. A significantly increased platelet 5-HT concentration in CD subjects, compared to controls, was lost after the correction according to the smoking status. Our results indicate an association of the studied HTR2A polymorphisms and their haplotypes with irritability and impulsivity traits, which may contribute to the aggressive and antisocial behavior in male adolescents with CD.
Collapse
Affiliation(s)
- Gordana Nedic Erjavec
- Rudjer Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10000 Zagreb, Croatia
| | - Lucija Tudor
- Rudjer Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10000 Zagreb, Croatia
| | - Matea Nikolac Perkovic
- Rudjer Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10000 Zagreb, Croatia
| | - Josip Podobnik
- Department of Psychiatry, Psychiatric Hospital for Children and Youth Zagreb, Kukuljeviceva 11, 10000 Zagreb, Croatia
| | - Katarina Dodig Curkovic
- Department for Child and Adolescent Psychiatry, Clinical Hospital Center Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Mario Curkovic
- Family Medicine, Park Kralja Petra Krešimira IV. 6, 31000 Osijek, Croatia
| | - Dubravka Svob Strac
- Rudjer Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10000 Zagreb, Croatia
| | | | - Marco Bortolato
- Department of Pharmacology & Toxicology, College of Pharmacy, Salt Lake City, 30 South 2000 East, UT 84112, USA.
| | - Nela Pivac
- Rudjer Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10000 Zagreb, Croatia.
| |
Collapse
|
6
|
Cristiano L. The pseudogenes of eukaryotic translation elongation factors (EEFs): Role in cancer and other human diseases. Genes Dis 2022; 9:941-958. [PMID: 35685457 PMCID: PMC9170609 DOI: 10.1016/j.gendis.2021.03.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/29/2021] [Indexed: 02/06/2023] Open
Abstract
The eukaryotic translation elongation factors (EEFs), i.e. EEF1A1, EEF1A2, EEF1B2, EEF1D, EEF1G, EEF1E1 and EEF2, are coding-genes that play a central role in the elongation step of translation but are often altered in cancer. Less investigated are their pseudogenes. Recently, it was demonstrated that pseudogenes have a key regulatory role in the cell, especially via non-coding RNAs, and that the aberrant expression of ncRNAs has an important role in cancer development and progression. The present review paper, for the first time, collects all that published about the EEFs pseudogenes to create a base for future investigations. For most of them, the studies are in their infancy, while for others the studies suggest their involvement in normal cell physiology but also in various human diseases. However, more investigations are needed to understand their functions in both normal and cancer cells and to define which can be useful biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Luigi Cristiano
- R&D Division, Prestige, 18 via Vecchia, Terranuova Bracciolini, AR 52028, Italy
| |
Collapse
|
7
|
Lata S, Mishra R, Arya RP, Arora P, Lahon A, Banerjea AC, Sood V. Where all the Roads Meet? A Crossover Perspective on Host Factors Regulating SARS-CoV-2 infection. J Mol Biol 2022; 434:167403. [PMID: 34914966 PMCID: PMC8666384 DOI: 10.1016/j.jmb.2021.167403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/18/2021] [Accepted: 12/07/2021] [Indexed: 01/11/2023]
Abstract
COVID-19 caused by SARS-CoV-2 is the latest pandemic which has thrown the world into an unprecedented social and economic uncertainties along with huge loss to humanity. Identification of the host factors regulating the replication of SARS-CoV-2 in human host may help in the development of novel anti-viral therapies to combat the viral infection and spread. Recently, some research groups used genome-wide CRISPR/Cas screening to identify the host factors critical for the SARS-CoV-2 replication and infection. A comparative analysis of these significant host factors (p < 0.05) identified fifteen proteins common in these studies. Apart from ACE2 (receptor for SARS-CoV-2 attachment), other common host factors were CSNK2B, GDI2, SLC35B2, DDX51, VPS26A, ARPP-19, C1QTNF7, ALG6, LIMA1, COG3, COG8, BCOR, LRRN2 and TLR9. Additionally, viral interactome of these host factors revealed that many of them were associated with several SARS-CoV-2 proteins as well. Interestingly, some of these host factors have already been shown to be critical for the pathogenesis of other viruses suggesting their crucial role in virus-host interactions. Here, we review the functions of these host factors and their role in other diseases with special emphasis on viral diseases.
Collapse
Affiliation(s)
- Sneh Lata
- Virology Laboratory, National Institute of Immunology, New Delhi, India
| | - Ritu Mishra
- Virology Laboratory, National Institute of Immunology, New Delhi, India
| | - Ravi P. Arya
- KSBS, Indian Institute of Technology, New Delhi, India
| | - Pooja Arora
- Hansraj College, University of Delhi, New Delhi, India
| | | | - Akhil C. Banerjea
- Institute of Advanced Virology, Kerala, India,Corresponding authors
| | - Vikas Sood
- Biochemistry Department, Jamia Hamdard, New Delhi, India,Corresponding authors
| |
Collapse
|
8
|
Chiocchetti AG, Yousaf A, Waltes R, Bernhard A, Martinelli A, Ackermann K, Haslinger D, Rotter B, Krezdorn N, Konrad K, Kohls G, Vetro A, Hervas A, Fernández-Rivas A, Freitag CM. The methylome in females with adolescent Conduct Disorder: Neural pathomechanisms and environmental risk factors. PLoS One 2022; 17:e0261691. [PMID: 35089926 PMCID: PMC8797262 DOI: 10.1371/journal.pone.0261691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 12/07/2021] [Indexed: 11/24/2022] Open
Abstract
Conduct Disorder (CD) is an impairing psychiatric disorder of childhood and adolescence characterized by aggressive and dissocial behavior. Environmental factors such as maternal smoking during pregnancy, socio-economic status, trauma, or early life stress are associated with CD. Although the number of females with CD is rising in Western societies, CD is under-researched in female cohorts. We aimed at exploring the epigenetic signature of females with CD and its relation to psychosocial and environmental risk factors. We performed HpaII sensitive genome-wide methylation sequencing of 49 CD girls and 50 matched typically developing controls and linear regression models to identify differentially methylated CpG loci (tags) and regions. Significant tags and regions were mapped to the respective genes and tested for enrichment in pathways and brain developmental processes. Finally, epigenetic signatures were tested as mediators for CD-associated risk factors. We identified a 12% increased methylation 5’ of the neurite modulator SLITRK5 (FDR = 0.0046) in cases within a glucocorticoid receptor binding site. Functionally, methylation positively correlated with gene expression in lymphoblastoid cell lines. At systems-level, genes (uncorr. P < 0.01) were associated with development of neurons, neurite outgrowth or neuronal developmental processes. At gene expression level, the associated gene-networks are activated perinatally and during early childhood in neocortical regions, thalamus and striatum, and expressed in amygdala and hippocampus. Specifically, the epigenetic signatures of the gene network activated in the thalamus during early childhood correlated with the effect of parental education on CD status possibly mediating its protective effect. The differential methylation patterns identified in females with CD are likely to affect genes that are expressed in brain regions previously indicated in CD. We provide suggestive evidence that protective effects are likely mediated by epigenetic mechanisms impairing specific brain developmental networks and therefore exerting a long-term effect on neural functions in CD. Our results are exploratory and thus, further replication is needed.
Collapse
Affiliation(s)
- Andreas G. Chiocchetti
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University Frankfurt, Frankfurt am Main, Germany
- * E-mail: ;
| | - Afsheen Yousaf
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Regina Waltes
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Anka Bernhard
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University Frankfurt, Frankfurt am Main, Germany
- * E-mail: ;
| | - Anne Martinelli
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Katharina Ackermann
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Denise Haslinger
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | | | - Kerstin Konrad
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Child Neuropsychology Section, University Hospital, RWTH Aachen, Aachen, Germany
- Molecular Neuroscience and Neuroimaging, Institute of Neuroscience and Medicine (INM-11) JARA BRAIN Institute II, Research Center Juelich, Juelich, Germany
| | - Gregor Kohls
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Child Neuropsychology Section, University Hospital, RWTH Aachen, Aachen, Germany
| | - Agnes Vetro
- Department of Pediatrics and Pediatrics Health Center, Child and Adolescent Psychiatry, Szeged, Szeged University, Szeged, Hungary
| | - Amaia Hervas
- Child and Adolescent Mental Health Service, Hospital Universitario Mutua de Terrassa, Barcelona, Spain
| | | | - Christine M. Freitag
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
9
|
Hay DF, Paine AL, Perra O, Cook KV, Hashmi S, Robinson C, Kairis V, Slade R. Prosocial and Aggressive Behavior: A Longitudinal Study. Monogr Soc Res Child Dev 2021; 86:7-103. [PMID: 33973244 PMCID: PMC9943493 DOI: 10.1111/mono.12427] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Developmental theorists have made strong claims about the fundamental prosocial or aggressive nature of the human infant. However, only rarely have prosocial behavior and aggression been studied together in the same sample. We charted the parallel development of both behaviors from infancy to childhood in a British community sample, using a two-construct, multimethod longitudinal design. Data were drawn from the Cardiff Child Development Study (CCDS), a prospective longitudinal study of a volunteer sample of parents and their firstborn children. A sample of 332 mothers was recruited from National Health Service (NHS) prenatal clinics and general practice clinics in Wales, UK, between Fall of 2005 and Summer of 2007. Potential participants represented the full range of sociodemographic classifications of neighborhoods. Participating families were divided about equally between middle- and working-class families, were somewhat more likely to have sons than daughters, and the majority (90%) were in a stable partnership. In response to standard categories recommended for use in Wales at the time, the majority (93%) of mothers reported themselves as Welsh, Scottish, English, or Irish; most others named a European or South Asian nationality. Of the 332 families agreeing to participate, 321 mothers (Mage = 28 years) and 285 partners (Mage = 31 years) were interviewed during the pregnancy and 321 of the families contributed data at least once after the child's birth. After an initial home visit at 6 months, data collection occurred in four additional waves of testing when children's mean ages were approximately 1, 1.5, 2.5, and 7 years. Data collection alternated between family homes and Cardiff University. Of those families seen after the child's birth, 89% were assessed at the final wave of testing. Data collection ended in 2015. Methods included direct observation, experimental tasks, and collection of reports from mothers, fathers, other relatives or family friends, and classroom teachers. Interactions with a familiar peer were observed at 1.5 years. Interactions with unfamiliar peers took place during experimental birthday parties at 1 and 2.5 years. At 7 years, parents were interviewed, parents and teachers completed questionnaires, and the children engaged in cognitive and social decision-making tasks. Based on reports from parents and other informants who knew the children well, individual differences in both prosocial behavior and aggression were evident in children. Both types of behavior showed stability across the second and third years. The association between prosocial behavior and aggression changed over time: at 1.5 years, they were not significantly related (the association approached zero), but they became negatively correlated by 3 years. Different patterns were seen when children played with familiar versus unfamiliar peers. At 1.5 years, when children were observed at home with a familiar peer, prosocial behavior and aggression were unrelated, thus showing a pattern of results like that seen in the analysis of informants' reports. However, a different pattern emerged during the experimental birthday parties with unfamiliar peers: prosocial behavior and aggression were positively correlated at both 1 and 2.5 years, contributing to a general sociability factor at both ages. Gender differences in prosocial behavior were evident in informants' reports and were also evident at the 1-year (though not the 2.5-year) birthday parties. In contrast, gender differences in both prosocial behavior and aggression were evident by 7 years, both in children's aggressive decision-making and in their parents' and teachers' reports of children's aggressive behavior at home and school. By age 7, children's aggressive decision-making and behavior were inversely associated with their verbal skills, working memory, and emotional understanding. Some children had developed aggressive behavioral problems and callous-unemotional traits. A few (12%) met diagnostic criteria for conduct disorder or oppositional-defiant disorders, which had been predicted by early angry aggressiveness and lack of empathy for other people. Taken together, the findings revealed a gradual disaggregation of two ways in which children interact with other people. Individual differences in both prosocial behavior and aggression revealed continuity over time, with gender differences emerging first in prosocial behavior, then in aggression. Restrictions in the participant sample and the catchment area (e.g., all were first-time parents; all were drawn from a single region in the United Kingdom) mean that it is not possible to generalize findings broadly. It will be important to expand the study of prosocial behavior and aggression in other family and environmental contexts in future work. Learning more about early appearing individual differences in children's approaches to the social world may be useful for both educational and clinical practice.
Collapse
Affiliation(s)
| | | | - Oliver Perra
- School of Nursing and Midwifery, Centre for Evidence and Social InnovationQueen's University Belfast
| | | | - Salim Hashmi
- Department of Psychology, Institute of Psychiatry, Psychology and NeuroscienceKing's College London
| | | | | | | |
Collapse
|
10
|
Lai D, Alipanahi B, Fontanillas P, Schwantes-An TH, Aasly J, Alcalay RN, Beecham GW, Berg D, Bressman S, Brice A, Brockman K, Clark L, Cookson M, Das S, Van Deerlin V, Follett J, Farrer MJ, Trinh J, Gasser T, Goldwurm S, Gustavsson E, Klein C, Lang AE, Langston JW, Latourelle J, Lynch T, Marder K, Marras C, Martin ER, McLean CY, Mejia-Santana H, Molho E, Myers RH, Nuytemans K, Ozelius L, Payami H, Raymond D, Rogaeva E, Rogers MP, Ross OA, Samii A, Saunders-Pullman R, Schüle B, Schulte C, Scott WK, Tanner C, Tolosa E, Tomkins JE, Vilas D, Trojanowski JQ, Uitti R, Vance JM, Visanji NP, Wszolek ZK, Zabetian CP, Mirelman A, Giladi N, Orr Urtreger A, Cannon P, Fiske B, Foroud T. Genomewide Association Studies of LRRK2 Modifiers of Parkinson's Disease. Ann Neurol 2021; 90:76-88. [PMID: 33938021 PMCID: PMC8252519 DOI: 10.1002/ana.26094] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 02/03/2023]
Abstract
Objective The aim of this study was to search for genes/variants that modify the effect of LRRK2 mutations in terms of penetrance and age‐at‐onset of Parkinson's disease. Methods We performed the first genomewide association study of penetrance and age‐at‐onset of Parkinson's disease in LRRK2 mutation carriers (776 cases and 1,103 non‐cases at their last evaluation). Cox proportional hazard models and linear mixed models were used to identify modifiers of penetrance and age‐at‐onset of LRRK2 mutations, respectively. We also investigated whether a polygenic risk score derived from a published genomewide association study of Parkinson's disease was able to explain variability in penetrance and age‐at‐onset in LRRK2 mutation carriers. Results A variant located in the intronic region of CORO1C on chromosome 12 (rs77395454; p value = 2.5E‐08, beta = 1.27, SE = 0.23, risk allele: C) met genomewide significance for the penetrance model. Co‐immunoprecipitation analyses of LRRK2 and CORO1C supported an interaction between these 2 proteins. A region on chromosome 3, within a previously reported linkage peak for Parkinson's disease susceptibility, showed suggestive associations in both models (penetrance top variant: p value = 1.1E‐07; age‐at‐onset top variant: p value = 9.3E‐07). A polygenic risk score derived from publicly available Parkinson's disease summary statistics was a significant predictor of penetrance, but not of age‐at‐onset. Interpretation This study suggests that variants within or near CORO1C may modify the penetrance of LRRK2 mutations. In addition, common Parkinson's disease associated variants collectively increase the penetrance of LRRK2 mutations. ANN NEUROL 2021;90:82–94
Collapse
Affiliation(s)
- Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | | | | | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Jan Aasly
- Department of Neurology, St. Olavs Hospital, Trondheim, Norway
| | - Roy N Alcalay
- Department of Neurology, Columbia University, New York, NY
| | - Gary W Beecham
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL
| | - Daniela Berg
- Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany.,Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Susan Bressman
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexis Brice
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, Inserm, CNRS, University Hospital Pitié-Salpêtrière, Paris, France
| | - Kathrin Brockman
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Lorraine Clark
- Department of Pathology and Cell Biology, Columbia University, New York, NY
| | - Mark Cookson
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD
| | | | - Vivianna Van Deerlin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jordan Follett
- Laboratory of Neurogenetics and Neuroscience, Fixel Institute for Neurological Diseases, McKnight Brain Institute, L5-101D, UF Clinical and Translational Science Institute, University of Florida, Gainesville, FL
| | - Matthew J Farrer
- Laboratory of Neurogenetics and Neuroscience, Fixel Institute for Neurological Diseases, McKnight Brain Institute, L5-101D, UF Clinical and Translational Science Institute, University of Florida, Gainesville, FL
| | - Joanne Trinh
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Thomas Gasser
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Emil Gustavsson
- Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada
| | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Anthony E Lang
- The Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Canada
| | - J William Langston
- Departments of Neurology, Neuroscience, and Pathology, Stanford University School of Medicine, Stanford, CA
| | | | - Timothy Lynch
- Dublin Neurological Institute at the Mater Misericordiae University Hospital, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Karen Marder
- Department of Neurology and Psychiatry, Taub Institute and Sergievsky Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Connie Marras
- The Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Canada
| | - Eden R Martin
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL
| | - Cory Y McLean
- 23andMe, Inc., Sunnyvale, CA.,Google LLC, Cambridge, MA
| | | | - Eric Molho
- Department of Neurology, Albany Medical College, Albany, NY
| | | | - Karen Nuytemans
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL
| | - Laurie Ozelius
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Haydeh Payami
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Deborah Raymond
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Neurology, University of Toronto, Toronto, Canada
| | - Michael P Rogers
- Department of General Surgery, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Owen A Ross
- Departments of Neuroscience and Clinical Genomics, Mayo Clinic, Jacksonville, FL.,School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Ali Samii
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, WA
| | | | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Claudia Schulte
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - William K Scott
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL
| | - Caroline Tanner
- University of California, San Francisco Veterans Affairs Health Care System, San Francisco, CA
| | - Eduardo Tolosa
- Parkinson Disease and Movement Disorders Unit, Hospital Clínic Universitari, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | | | - Dolores Vilas
- Parkinson Disease and Movement Disorders Unit, Hospital Clínic Universitari, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | -
- 23andMe, Inc., Sunnyvale, CA
| | - Ryan Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL
| | - Jeffery M Vance
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL
| | - Naomi P Visanji
- The Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Canada
| | | | - Cyrus P Zabetian
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, WA
| | - Anat Mirelman
- Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Nir Giladi
- Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Avi Orr Urtreger
- Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | - Brian Fiske
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
11
|
Demontis D, Walters RK, Rajagopal VM, Waldman ID, Grove J, Als TD, Dalsgaard S, Ribasés M, Bybjerg-Grauholm J, Bækvad-Hansen M, Werge T, Nordentoft M, Mors O, Mortensen PB, Cormand B, Hougaard DM, Neale BM, Franke B, Faraone SV, Børglum AD. Risk variants and polygenic architecture of disruptive behavior disorders in the context of attention-deficit/hyperactivity disorder. Nat Commun 2021; 12:576. [PMID: 33495439 PMCID: PMC7835232 DOI: 10.1038/s41467-020-20443-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 11/30/2020] [Indexed: 11/29/2022] Open
Abstract
Attention-Deficit/Hyperactivity Disorder (ADHD) is a childhood psychiatric disorder often comorbid with disruptive behavior disorders (DBDs). Here, we report a GWAS meta-analysis of ADHD comorbid with DBDs (ADHD + DBDs) including 3802 cases and 31,305 controls. We identify three genome-wide significant loci on chromosomes 1, 7, and 11. A meta-analysis including a Chinese cohort supports that the locus on chromosome 11 is a strong risk locus for ADHD + DBDs across European and Chinese ancestries (rs7118422, P = 3.15×10−10, OR = 1.17). We find a higher SNP heritability for ADHD + DBDs (h2SNP = 0.34) when compared to ADHD without DBDs (h2SNP = 0.20), high genetic correlations between ADHD + DBDs and aggressive (rg = 0.81) and anti-social behaviors (rg = 0.82), and an increased burden (polygenic score) of variants associated with ADHD and aggression in ADHD + DBDs compared to ADHD without DBDs. Our results suggest an increased load of common risk variants in ADHD + DBDs compared to ADHD without DBDs, which in part can be explained by variants associated with aggressive behavior. ADHD is often found to be comorbid with disruptive behavior disorders, but the genetic loci underlying this comorbidity are unknown. Here, the authors have performed a GWAS meta-analysis of ADHD with disruptive behavior disorders, finding three genome-wide significant loci in Europeans, and replicating one in a Chinese cohort.
Collapse
Affiliation(s)
- Ditte Demontis
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark. .,Center for Genomics and Personalized Medicine, Aarhus, Denmark. .,Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark.
| | - Raymond K Walters
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Veera M Rajagopal
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.,Center for Genomics and Personalized Medicine, Aarhus, Denmark.,Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Irwin D Waldman
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Jakob Grove
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.,Center for Genomics and Personalized Medicine, Aarhus, Denmark.,Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark.,Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Thomas D Als
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.,Center for Genomics and Personalized Medicine, Aarhus, Denmark.,Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
| | - Søren Dalsgaard
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Marta Ribasés
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain.,Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Catalonia, Spain.,Biomedical Network Research Center on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.,Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Jonas Bybjerg-Grauholm
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.,Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Maria Bækvad-Hansen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.,Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Thomas Werge
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.,GLOBE Institute, Center for GeoGenetics, University of Copenhagen, Copenhagen, Denmark.,Institute of Biological Psychiatry, MHC Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Merete Nordentoft
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.,Copenhagen University Hospital, Mental Health Centre Copenhagen Mental Health Services in the Capital Region of Denmark, Hellerup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ole Mors
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.,Psychosis Research Unit, Aarhus University Hospital, Aarhus, Denmark
| | - Preben Bo Mortensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.,Center for Genomics and Personalized Medicine, Aarhus, Denmark.,National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark.,Centre for Integrated Register-Based Research, Aarhus University, Aarhus, Denmark
| | | | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain.,Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues de Llobregat, Barcelona, Catalonia, Spain
| | - David M Hougaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.,Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Barbara Franke
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Anders D Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark. .,Center for Genomics and Personalized Medicine, Aarhus, Denmark. .,Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
12
|
Detention in Juvenile Correctional Facilities Is Associated with Higher Platelet Monoamine Oxidase B Activity in Males. Biomolecules 2020; 10:biom10111555. [PMID: 33203099 PMCID: PMC7697475 DOI: 10.3390/biom10111555] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/06/2020] [Accepted: 11/13/2020] [Indexed: 01/16/2023] Open
Abstract
Juvenile delinquency is related to several biological factors, yet very few vulnerability biomarkers have been identified. Previous data suggest that the enzyme monoamine oxidase B (MAO-B) influences several personality traits linked to the propensity to engage in delinquent behavior. Building on this evidence, we assessed whether conduct disorder (CD), juvenile delinquency adjudications, or detention in a correctional facility were associated with either platelet MAO-B activity or the MAOB rs1799836 polymorphism. The study enrolled 289 medication-free male youths, including 182 individuals detained in a correctional facility (with or without a diagnosis of CD). Of the remaining 107 participants, 26 subjects had a diagnosis of CD, and 81 were mentally healthy controls. Platelet MAO-B activity was determined by spectrophotofluorometry, while MAOB rs1799836 was genotyped using qPCR. Platelet MAO-B activity, corrected for age and smoking, was significantly higher in juvenile detainees (p < 0.001), irrespective of CD diagnosis. MAOB rs1799836 was not associated with platelet MAO-B activity or with detention in a correctional facility, CD diagnosis, or delinquent behavior. These data suggest that detention in a juvenile correctional facility increases platelet MAO-B activity in male adolescents. Future studies are needed to determine the mechanisms and functional significance of MAO-B peripheral elevation in juvenile male detainees.
Collapse
|
13
|
Jiang Y, Ming Q, Gao Y, Dong D, Sun X, Zhang X, Situ W, Yao S, Rao H. Effects of BDNF Val66Met polymorphisms on brain structures and behaviors in adolescents with conduct disorder. Eur Child Adolesc Psychiatry 2020; 29:479-488. [PMID: 31264106 DOI: 10.1007/s00787-019-01363-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 06/24/2019] [Indexed: 10/26/2022]
Abstract
Accumulating evidence suggests that neural abnormalities in conduct disorder (CD) may be subject to genetic influences, but few imaging studies have taken genetic variants into consideration. The Val66Met polymorphism of brain-derived neurotrophic factor (BDNF) has emerged as a high-interest genetic variant due to its importance in cortical maturation, and several studies have implicated its involvement in neurodevelopmental disorders. Thus, it is unclear how this polymorphism may influence brain anatomy and aberrant behaviors in CD. A total of 65 male adolescents with CD and 69 gender-, IQ- and socioeconomic status-matched healthy controls (HCs) (age range 13-17 years) were enrolled in this study. Analyses of variance (ANOVAs) were used to assess the main effects of CD diagnosis, BDNF genotype, and diagnosis-genotype interactions on brain anatomy and behaviors. We detected a significant main effect of BDNF genotype on temporal gyrification and antisocial behaviors, but not on CD symptoms. Diagnosis-genotype interactive effects were found for cortical thickness of the superior temporal and adjacent areas. These results suggest that the BDNF Val66Met polymorphism may exert its influence both on neural alterations and delinquent behaviors in CD patients. This initial evidence highlights the importance of elucidating potentially different pathways between BDNF genotype and cortical alterations or delinquent behaviors in CD patients.
Collapse
Affiliation(s)
- Yali Jiang
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, No. 139, Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China.,Center for Studies of Psychological Application, School of Psychology, South China Normal University, Guangzhou, Guangdong, People's Republic of China
| | - Qingsen Ming
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, No. 139, Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
| | - Yidian Gao
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, No. 139, Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
| | - Daifeng Dong
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, No. 139, Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
| | - Xiaoqiang Sun
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, No. 139, Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
| | - Xiaocui Zhang
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, No. 139, Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
| | - Weijun Situ
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Shuqiao Yao
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, No. 139, Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China. .,National Clinical Research Center on Psychiatry and Psychology, Changsha, Hunan, People's Republic of China. .,Medical Psychological Institute of Central South University, Changsha, Hunan, People's Republic of China.
| | - Hengyi Rao
- Center of Functional Neuroimaging, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
14
|
Genomics of human aggression: current state of genome-wide studies and an automated systematic review tool. Psychiatr Genet 2020; 29:170-190. [PMID: 31464998 DOI: 10.1097/ypg.0000000000000239] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
There are substantial differences, or variation, between humans in aggression, with its molecular genetic basis mostly unknown. This review summarizes knowledge on the genetic contribution to variation in aggression with the following three foci: (1) a comprehensive overview of reviews on the genetics of human aggression, (2) a systematic review of genome-wide association studies (GWASs), and (3) an automated tool for the selection of literature based on supervised machine learning. The phenotype definition 'aggression' (or 'aggressive behaviour', or 'aggression-related traits') included anger, antisocial behaviour, conduct disorder, and oppositional defiant disorder. The literature search was performed in multiple databases, manually and using a novel automated selection tool, resulting in 18 reviews and 17 GWASs of aggression. Heritability estimates of aggression in children and adults are around 50%, with relatively small fluctuations around this estimate. In 17 GWASs, 817 variants were reported as suggestive (P ≤ 1.0E), including 10 significant associations (P ≤ 5.0E). Nominal associations (P ≤ 1E) were found in gene-based tests for genes involved in immune, endocrine, and nervous systems. Associations were not replicated across GWASs. A complete list of variants and their position in genes and chromosomes are available online. The automated literature search tool produced literature not found by regular search strategies. Aggression in humans is heritable, but its genetic basis remains to be uncovered. No sufficiently large GWASs have been carried out yet. With increases in sample size, we expect aggression to behave like other complex human traits for which GWAS has been successful.
Collapse
|
15
|
Jager A, Amiri H, Bielczyk N, van Heukelum S, Heerschap A, Aschrafi A, Poelmans G, Buitelaar JK, Kozicz T, Glennon JC. Cortical control of aggression: GABA signalling in the anterior cingulate cortex. Eur Neuropsychopharmacol 2020; 30:5-16. [PMID: 29274996 DOI: 10.1016/j.euroneuro.2017.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 11/14/2017] [Accepted: 12/02/2017] [Indexed: 11/28/2022]
Abstract
Reduced top-down control by cortical areas is assumed to underlie pathological forms of aggression. While the precise underlying molecular mechanisms are still elusive, it seems that balancing the excitatory and inhibitory tones of cortical brain areas has a role in aggression control. The molecular mechanisms underpinning aggression control were examined in the BALB/cJ mouse model. First, these mice were extensively phenotyped for aggression and anxiety in comparison to BALB/cByJ controls. Microarray data was then used to construct a molecular landscape, based on the mRNAs that were differentially expressed in the brains of BALB/cJ mice. Subsequently, we provided corroborating evidence for the key findings from the landscape through 1H-magnetic resonance imaging and quantitative polymerase chain reactions, specifically in the anterior cingulate cortex (ACC). The molecular landscape predicted that altered GABA signalling may underlie the observed increased aggression and anxiety in BALB/cJ mice. This was supported by a 40% reduction of 1H-MRS GABA levels and a 20-fold increase of the GABA-degrading enzyme Abat in the ventral ACC. As a possible compensation, Kcc2, a potassium-chloride channel involved in GABA-A receptor signalling, was found increased. Moreover, we observed aggressive behaviour that could be linked to altered expression of neuroligin-2, a membrane-bound cell adhesion protein that mediates synaptogenesis of mainly inhibitory synapses. In conclusion, Abat and Kcc2 seem to be involved in modulating aggressive and anxious behaviours observed in BALB/cJ mice through affecting GABA signalling in the ACC.
Collapse
Affiliation(s)
- Amanda Jager
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| | - Houshang Amiri
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands; Neuroscience Research Centre, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Natalia Bielczyk
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Sabrina van Heukelum
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Armaz Aschrafi
- Laboratory of Molecular Biology, National Institute of Mental Health, Bethesda, United States
| | - Geert Poelmans
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, Nijmegen, The Netherlands
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Tamas Kozicz
- Department of Anatomy, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jeffrey C Glennon
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Nitric oxide synthase genotype interacts with stressful life events to increase aggression in male subjects in a population-representative sample. Eur Neuropsychopharmacol 2020; 30:56-65. [PMID: 31405541 DOI: 10.1016/j.euroneuro.2019.07.241] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 06/14/2019] [Accepted: 07/27/2019] [Indexed: 01/02/2023]
Abstract
Nitric oxide signalling has been implicated in impulsive and aggressive traits and behaviours in both animals and humans. In the present study, we investigated the effects of a functional variable number of tandem repeats (VNTR) polymorphism in exon 1f (ex1f) of the nitric oxide synthase 1 (NOS1) gene (NOS1 ex1f-VNTR) and stressful life events on aggressive behaviour in population representative sample of adolescents followed up from third grade to 25 years of age. We studied the younger cohort of the longitudinal Estonian Children Personality, Behaviour and Health Study (subjects in the last study wave n = 437, males n = 193; mean age 24.8 ± 0.5 years). Aggressive behaviour was rated at age 25 with the Illinois Bully Scale and Buss-Perry Aggression Questionnaire. Life history of aggression was evaluated in a structured interview. Stressful life events and family relationships were self-reported at age 15. The hypothesized risk genotype (homozygosity for the short allele) was associated with higher levels of aggression in males (statistical significance withstanding the multiple correction procedure). Exposure to stressful life events or adverse family relationships was associated with increased aggressive behaviour in subjects homozygous for either of the alleles, and these associations were mostly observed in males. However, these associations in these stratified analyses did not survive correction for multiple testing. Aggressiveness was relatively unaffected by the NOS1 ex1f-VNTR genotype in the female subjects even when taking exposure to childhood adversity into account. Our findings support the hypothesized involvement of a functional NOS1 polymorphism on aggression in a population representative sample of young adults.
Collapse
|
17
|
Vyas S, Constantino JN, Baldridge D. 22q11.2 duplication: a review of neuropsychiatric correlates and a newly observed case of prototypic sociopathy. Cold Spring Harb Mol Case Stud 2019; 5:mcs.a004291. [PMID: 31836587 PMCID: PMC6913156 DOI: 10.1101/mcs.a004291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 09/26/2019] [Indexed: 12/18/2022] Open
Abstract
Callous-unemotional (CU) traits are highly disabling behavioral characteristics, common predictors of delinquency and criminality, and pathognomonic for antisocial personality disorder. They are highly heritable, but their specific molecular genetic causes are unknown. Here, we briefly review the literature on neuropsychiatric correlates of 22q11.2 duplication and describe a newly identified case of a 737-kb microduplication within the low copy repeat (LCR) B-D region, involving a 13-yr-old early adoptee with mild developmental delay and severe, chronic antisocial behavior of early childhood onset. When psychiatric symptoms have been reported in relation to duplications in this specific region, 19% of the reports feature aggression—but never previously CU traits—as a component of the phenotype. We discuss the potential implications of gain of function in this chromosomal region for heritable origins of sociopathy and their possible relation to genetic influences on aggression.
Collapse
Affiliation(s)
- Sonam Vyas
- St. Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - John N Constantino
- Division of Child Psychiatry, Department of Psychiatry, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Dustin Baldridge
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
18
|
Camilleri M, Sandler RS, Peery AF. Etiopathogenetic Mechanisms in Diverticular Disease of the Colon. Cell Mol Gastroenterol Hepatol 2019; 9:15-32. [PMID: 31351939 PMCID: PMC6881605 DOI: 10.1016/j.jcmgh.2019.07.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 02/08/2023]
Abstract
This article reviews epidemiological evidence of heritability and putative mechanisms in diverticular disease, with greatest attention to 3 recent studies of genetic associations with diverticular disease based on genome-wide or whole-genome sequencing studies in large patient cohorts. We provide an analysis of the biological plausibility of the significant associations with gene variants reported and highlight the relevance of ANO1, CPI-17 (aka PPP1R14A), COLQ6, COL6A1, CALCB or CALCA, COL6A1, ARHGAP15, and S100A10 to colonic neuromuscular function and tissue properties that may result in altered compliance and predispose to the development of diverticular disease. Such studies also identify candidate genes for future studies.
Collapse
Affiliation(s)
- Michael Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Robert S Sandler
- Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Anne F Peery
- Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
19
|
Fairchild G, Hawes DJ, Frick PJ, Copeland WE, Odgers CL, Franke B, Freitag CM, De Brito SA. Conduct disorder. Nat Rev Dis Primers 2019; 5:43. [PMID: 31249310 DOI: 10.1038/s41572-019-0095-y] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/17/2019] [Indexed: 02/06/2023]
Abstract
Conduct disorder (CD) is a common and highly impairing psychiatric disorder that usually emerges in childhood or adolescence and is characterized by severe antisocial and aggressive behaviour. It frequently co-occurs with attention-deficit/hyperactivity disorder (ADHD) and often leads to antisocial personality disorder in adulthood. CD affects ~3% of school-aged children and is twice as prevalent in males than in females. This disorder can be subtyped according to age at onset (childhood-onset versus adolescent-onset) and the presence or absence of callous-unemotional traits (deficits in empathy and guilt). The aetiology of CD is complex, with contributions of both genetic and environmental risk factors and different forms of interplay among the two (gene-environment interaction and correlation). In addition, CD is associated with neurocognitive impairments; smaller grey matter volume in limbic regions such as the amygdala, insula and orbitofrontal cortex, and functional abnormalities in overlapping brain circuits responsible for emotion processing, emotion regulation and reinforcement-based decision-making have been reported. Lower hypothalamic-pituitary-adrenal axis and autonomic reactivity to stress has also been reported. Management of CD primarily involves parent-based or family-based psychosocial interventions, although stimulants and atypical antipsychotics are sometimes used, especially in individuals with comorbid ADHD.
Collapse
Affiliation(s)
| | - David J Hawes
- School of Psychology, University of Sydney, Sydney, New South Wales, Australia
| | - Paul J Frick
- Department of Psychology, Louisiana State University, Baton Rouge, LA, USA and Institute for Learning Science and Teacher Education, Australian Catholic University, Brisbane, Queensland, Australia
| | | | - Candice L Odgers
- Department of Psychological Science, School of Social Ecology, University of California, Irvine, CA, USA
| | - Barbara Franke
- Departments of Human Genetics and Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Christine M Freitag
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Stephane A De Brito
- School of Psychology and Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
20
|
Hollister BM, Farber-Eger E, Aldrich MC, Crawford DC. A Social Determinant of Health May Modify Genetic Associations for Blood Pressure: Evidence From a SNP by Education Interaction in an African American Population. Front Genet 2019; 10:428. [PMID: 31134134 PMCID: PMC6523518 DOI: 10.3389/fgene.2019.00428] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/18/2019] [Indexed: 01/11/2023] Open
Abstract
African Americans experience the highest burden of hypertension in the United States compared with other groups. Genetic contributions to this complex condition are now emerging in this as well as other populations through large-scale genome-wide association studies (GWAS) and meta-analyses. Despite these recent discovery efforts, relatively few large-scale studies of blood pressure have considered the joint influence of genetics and social determinants of health despite extensive evidence supporting their impact on hypertension. To identify these expected interactions, we accessed a subset of the Vanderbilt University Medical Center (VUMC) biorepository linked to de-identified electronic health records (EHRs) of adult African Americans genotyped using the Illumina Metabochip (n = 2,577). To examine potential interactions between education, a recognized social determinant of health, and genetic variants contributing to blood pressure, we used linear regression models to investigate two-way interactions for systolic and diastolic blood pressure (DBP). We identified a two-way interaction between rs6687976 and education affecting DBP (p = 0.052). Individuals homozygous for the minor allele and having less than a high school education had higher DBP compared with (1) individuals homozygous for the minor allele and high school education or greater and (2) individuals not homozygous for the minor allele and less than a high school education. To our knowledge, this is the first EHR -based study to suggest a gene-environment interaction for blood pressure in African Americans, supporting the hypothesis that genetic contributions to hypertension may be modulated by social factors.
Collapse
Affiliation(s)
- Brittany M Hollister
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Eric Farber-Eger
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Melinda C Aldrich
- Department of Thoracic Surgery, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Dana C Crawford
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
21
|
Wozniak J, Wilens T, DiSalvo M, Farrell A, Wolenski R, Faraone SV, Biederman J. Comorbidity of bipolar I disorder and conduct disorder: a familial risk analysis. Acta Psychiatr Scand 2019; 139:361-368. [PMID: 30758064 PMCID: PMC6476307 DOI: 10.1111/acps.13013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2019] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To investigate the association between pediatric bipolar I (BP-I) disorder and conduct disorder (CD) using familial risk analysis. METHOD We compared diagnoses in relatives of youth in four proband groups defined by the presence or absence of BP-I and CD: (1) probands with neither CD nor BP-I (probands: N = 550; relatives: N = 1656), (2) probands with CD and without BP-I (probands: N = 40; relatives: N = 127), (3) probands with BP-I and without CD (probands: N = 197; relatives: N = 579), and (4) probands with both CD and BP-I (probands: N = 176; relatives: N = 488). All subjects were evaluated with structured diagnostic interviews, and diagnoses of relatives were made blind to the diagnoses of probands. RESULTS Relatives of probands with BP-I disorder had high rates of BP-I, and relatives of probands with CD had high rates of CD irrespective of the comorbidity with the other disorder. Relatives of probands with the combined condition of CD and BP-I had high rates of the combined condition. CONCLUSION The finding of cosegregation between BP-I disorder and CD is consistent with the hypothesis that the combined condition represents a distinct subtype of either disorder.
Collapse
Affiliation(s)
- Janet Wozniak
- Pediatric Psychopharmacology Program, Division of Child
Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Psychiatry, Massachusetts General Hospital
and Harvard Medical School, Boston, MA 02114, USA
| | - Timothy Wilens
- Pediatric Psychopharmacology Program, Division of Child
Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Psychiatry, Massachusetts General Hospital
and Harvard Medical School, Boston, MA 02114, USA
| | - Maura DiSalvo
- Pediatric Psychopharmacology Program, Division of Child
Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Abigail Farrell
- Pediatric Psychopharmacology Program, Division of Child
Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Rebecca Wolenski
- Pediatric Psychopharmacology Program, Division of Child
Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stephen V. Faraone
- Department of Psychiatry and Behavioral Sciences, SUNY
Upstate Medical University, Syracuse, New York, USA
| | - Joseph Biederman
- Pediatric Psychopharmacology Program, Division of Child
Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Psychiatry, Massachusetts General Hospital
and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
22
|
Musci RJ, Bettencourt AF, Sisto D, Maher B, Masyn K, Ialongo NS. Violence exposure in an urban city: A GxE interaction with aggressive and impulsive behaviors. J Child Psychol Psychiatry 2019; 60:72-81. [PMID: 30159911 PMCID: PMC6392042 DOI: 10.1111/jcpp.12966] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/23/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Previous research has demonstrated a reciprocal relationship between community violence exposure and disruptive behavior problems among youth. No study to date, however, has explored the potential interaction between violence exposure in early adolescence and genetics. METHODS We explore the gene x environment interaction's impact on teacher-rated aggressive and impulsive behaviors. Violence exposure during the middle school years was assessed using self-report. Genetic data collection occurred in emerging adulthood. A polygenic score was created using findings from a conduct disorder symptomatology genome-wide association study. RESULTS Three longitudinal classes of teacher reported aggressive and impulsive behavior were identified. We found a significant relationship between violence exposure and class membership. There was a significant GxE interaction, such that those with below average levels of the polygenic score and who were exposed to violence were more likely to be in the moderately high aggressive and impulsive class as compared to the no to low class. CONCLUSIONS These findings highlight the influence of genetic risk together with violence exposure on adolescent problem behavior. Although youth may have little control over the environments in which they live, interventions can and should focus on helping all youth.
Collapse
Affiliation(s)
- Rashelle J. Musci
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Amie F. Bettencourt
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD
| | - Danielle Sisto
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Brion Maher
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Katherine Masyn
- Division of Epidemiology & Biostatistics, Georgia State University School of Public Health, Atlanta, GA, USA
| | - Nicholas S. Ialongo
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
23
|
Rabinowitz JA, Musci RJ, Milam AJ, Benke K, Uhl GR, Sisto DY, Ialongo NS, Maher BS. The interplay between externalizing disorders polygenic risk scores and contextual factors on the development of marijuana use disorders. Drug Alcohol Depend 2018; 191:365-373. [PMID: 30195949 PMCID: PMC8005265 DOI: 10.1016/j.drugalcdep.2018.07.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 07/13/2018] [Accepted: 07/18/2018] [Indexed: 12/28/2022]
Abstract
Externalizing disorders have been extensively linked to substance use problems. However, less is known about whether genetic factors underpinning externalizing disorders and environmental features interact to predict substance use disorders (i.e., marijuana abuse and dependence) among urban African Americans. We examined whether polygenic risk scores (PRS) for conduct disorder (CD) and attention-deficit hyperactivity disorder (ADHD) interacted with contextual factors (i.e., parental monitoring, community disadvantage) to influence risk for marijuana use disorders in a sample of African American youth. Participants (N=1,050; 44.2% male) were initially recruited for an elementary school-based universal prevention trial in a Mid-Atlantic city and followed through age 20. Participants reported on their parental monitoring in sixth grade and whether they were diagnosed with marijuana abuse or dependence at age 20. Blood or saliva samples were genotyped using the Affymetrix 6.0 microarrays. The CD and ADHD PRS were created based on genome-wide association studies conducted by Dick et al. (2010) and Demontis et al. (2017), respectively. Community disadvantage was calculated based on census data when participants were in sixth grade. There was an interaction between the CD PRS and community disadvantage such that a higher CD PRS was associated with greater risk for a marijuana use disorder at higher levels of neighborhood disadvantage. This finding should be interpreted with caution owing to the number of significance tests performed. Implications for etiological models and future research directions are presented.
Collapse
Affiliation(s)
- Jill A Rabinowitz
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 624 N. Broadway, Baltimore, MD 21205, United States.
| | - Rashelle J Musci
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 624 N. Broadway, Baltimore, MD 21205, United States
| | - Adam J Milam
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 624 N. Broadway, Baltimore, MD 21205, United States
| | - Kelly Benke
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 624 N. Broadway, Baltimore, MD 21205, United States
| | - George R Uhl
- New Mexico VA Healthcare System, 1501 San Pedro Drive, SE, Albuquerque, NM, 87108 United States
| | - Danielle Y Sisto
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 624 N. Broadway, Baltimore, MD 21205, United States
| | - Nicholas S Ialongo
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 624 N. Broadway, Baltimore, MD 21205, United States
| | - Brion S Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 624 N. Broadway, Baltimore, MD 21205, United States
| |
Collapse
|
24
|
Conduct disorder in adolescent females: current state of research and study design of the FemNAT-CD consortium. Eur Child Adolesc Psychiatry 2018; 27:1077-1093. [PMID: 29948230 DOI: 10.1007/s00787-018-1172-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 05/22/2018] [Indexed: 01/09/2023]
Abstract
Conduct disorder (CD) is a common and highly impairing psychiatric disorder of childhood and adolescence that frequently leads to poor physical and mental health outcomes in adulthood. The prevalence of CD is substantially higher in males than females, and partly due to this, most research on this condition has used all-male or predominantly male samples. Although the number of females exhibiting CD has increased in recent decades, the majority of studies on neurobiological measures, neurocognitive phenotypes, and treatments for CD have focused on male subjects only, despite strong evidence for sex differences in the aetiology and neurobiology of CD. Here, we selectively review the existing literature on CD and related phenotypes in females, focusing in particular on sex differences in CD symptoms, patterns of psychiatric comorbidity, and callous-unemotional personality traits. We also consider studies investigating the neurobiology of CD in females, with a focus on studies using genetic, structural and functional neuroimaging, psychophysiological, and neuroendocrinological methods. We end the article by providing an overview of the study design of the FemNAT-CD consortium, an interdisciplinary, multi-level and multi-site study that explicitly focuses on CD in females, but which is also investigating sex differences in the causes, developmental course, and neurobiological correlates of CD.
Collapse
|
25
|
Gard AM, Dotterer HL, Hyde LW. Genetic influences on antisocial behavior: recent advances and future directions. Curr Opin Psychol 2018; 27:46-55. [PMID: 30145531 DOI: 10.1016/j.copsyc.2018.07.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/24/2018] [Accepted: 07/30/2018] [Indexed: 12/24/2022]
Abstract
Understanding the etiology of antisocial behavior (i.e. violence, criminality, rule-breaking), is essential to the development of more effective prevention and intervention strategies. We provide a summary of the genetic correlates of antisocial behavior, drawing upon findings from behavioral, molecular, and statistical genetics. Across methodologies, our review highlights the centrality of environmental moderators of genetic effects, and how behavioral heterogeneity in antisocial behavior is an important consideration for genetic studies. We also review novel analytic techniques and neurogenetic approaches that can be used to examine how genetic variation predicts antisocial behavior. Finally, to illustrate how findings may converge across approaches, we describe pathways from genetic variability in oxytocin signaling to subtypes of antisocial behavior.
Collapse
Affiliation(s)
- Arianna M Gard
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Hailey L Dotterer
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Luke W Hyde
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA; Center for Human Growth and Development, University of Michigan, Ann Arbor, MI, USA; Survey Research Center of the Institute for Social Research, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
26
|
Holz NE, Zohsel K, Laucht M, Banaschewski T, Hohmann S, Brandeis D. Gene x environment interactions in conduct disorder: Implications for future treatments. Neurosci Biobehav Rev 2018; 91:239-258. [DOI: 10.1016/j.neubiorev.2016.08.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 07/27/2016] [Accepted: 08/15/2016] [Indexed: 01/30/2023]
|
27
|
Salvatore JE, Dick DM. Genetic influences on conduct disorder. Neurosci Biobehav Rev 2018; 91:91-101. [PMID: 27350097 PMCID: PMC5183514 DOI: 10.1016/j.neubiorev.2016.06.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 05/22/2016] [Accepted: 06/22/2016] [Indexed: 01/08/2023]
Abstract
Conduct disorder (CD) is a moderately heritable psychiatric disorder of childhood and adolescence characterized by aggression toward people and animals, destruction of property, deceitfulness or theft, and serious violation of rules. Genome-wide scans using linkage and association methods have identified a number of suggestive genomic regions that are pending replication. A small number of candidate genes (e.g., GABRA2, MAOA, SLC6A4, AVPR1A) are associated with CD related phenotypes across independent studies; however, failures to replicate also exist. Studies of gene-environment interplay show that CD genetic predispositions also contribute to selection into higher-risk environments, and that environmental factors can alter the importance of CD genetic factors and differentially methylate CD candidate genes. The field's understanding of CD etiology will benefit from larger, adequately powered studies in gene identification efforts; the incorporation of polygenic approaches in gene-environment interplay studies; attention to the mechanisms of risk from genes to brain to behavior; and the use of genetically informative data to test quasi-causal hypotheses about purported risk factors.
Collapse
Affiliation(s)
- Jessica E Salvatore
- Department of Psychology and the Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, VCU PO Box 842018, 806 West Franklin Street, Richmond, VA 23284-2018, USA.
| | - Danielle M Dick
- Department of Psychology, African American Studies, and Human & Molecular Genetics, VCU PO Box 842509, Richmond, VA 23284-2509, USA
| |
Collapse
|
28
|
Macrì S, Zoratto F, Chiarotti F, Laviola G. Can laboratory animals violate behavioural norms? Towards a preclinical model of conduct disorder. Neurosci Biobehav Rev 2018; 91:102-111. [DOI: 10.1016/j.neubiorev.2017.01.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/15/2016] [Accepted: 01/18/2017] [Indexed: 11/25/2022]
|
29
|
Ruisch IH, Buitelaar JK, Glennon JC, Hoekstra PJ, Dietrich A. Pregnancy risk factors in relation to oppositional-defiant and conduct disorder symptoms in the Avon Longitudinal Study of Parents and Children. J Psychiatr Res 2018; 101:63-71. [PMID: 29550610 DOI: 10.1016/j.jpsychires.2018.02.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 02/19/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pregnancy factors have been implicated in offspring oppositional-defiant disorder (ODD) and conduct disorder (CD) symptoms. Literature still holds notable limitations, such as studying only a restricted set of pregnancy factors, use of screening questionnaires which assess broadly defined outcome measures, and lack of control for disruptive behavior comorbidity and genetic confounds. We aimed to address these gaps by prospectively studying a broad range of pregnancy factors in relation to both offspring ODD and CD symptomatology in the Avon Longitudinal Study of Parent and Children. METHODS Outcomes were ODD and CD symptom scores at age 7;9 years using the Development and Well-Being Assessment interview. We analyzed maternal (N ≈ 6300) and teacher ratings (N ≈ 4400) of ODD and CD scores separately using negative binomial regression in multivariable models. Control variables included comorbid attention-deficit/hyperactivity disorder symptoms, ODD or CD symptoms as appropriate, and genetic risk scores based on an independent CD genome-wide association study. RESULTS Higher ODD symptom scores were linked to paracetamol use (IRR = 1.24 [98.3% confidence interval 1.05-1.47], P = 0.002, teacher ratings) and life events stress (IRR = 1.22 [1.07-1.39], P = 0.002, maternal ratings) during pregnancy. Higher CD symptom scores were linked to maternal smoking (IRR = 1.33 [1.18-1.51], P < 0.001, maternal ratings), life events stress (IRR = 1.24 [1.11-1.38], P < 0.001, maternal ratings) and depressive symptoms (IRR = 1.14 [1.01-1.30], P = 0.006, maternal ratings) during pregnancy. CONCLUSIONS Common and potentially preventable pregnancy risk factors were independently related to both offspring ODD and CD symptomatology in children from the general population. Future studies should further address genetic confounds and confounding by environmental factors later in life.
Collapse
Affiliation(s)
- I Hyun Ruisch
- University of Groningen, University Medical Center Groningen, Department of Child and Adolescent Psychiatry, Hanzeplein 1, 9713GZ Groningen, The Netherlands.
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands; Karakter Child and Adolescent Psychiatry University Centre, Reinier Postlaan 12, 6525 GC Nijmegen, The Netherlands.
| | - Jeffrey C Glennon
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands.
| | - Pieter J Hoekstra
- University of Groningen, University Medical Center Groningen, Department of Child and Adolescent Psychiatry, Hanzeplein 1, 9713GZ Groningen, The Netherlands.
| | - Andrea Dietrich
- University of Groningen, University Medical Center Groningen, Department of Child and Adolescent Psychiatry, Hanzeplein 1, 9713GZ Groningen, The Netherlands.
| |
Collapse
|
30
|
Musci RJ, Bettencourt AF, Sisto D, Maher B, Uhl G, Ialongo N, Bradshaw CP. Evaluating the genetic susceptibility to peer reported bullying behaviors. Psychiatry Res 2018; 263:193-198. [PMID: 29573659 PMCID: PMC6085882 DOI: 10.1016/j.psychres.2018.03.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 10/17/2022]
Abstract
Bullying is a significant public health concern with lasting impacts on youth. Although environmental risk factors for bullying have been well-characterized, genetic influences on bullying are not well understood. This study explored the role of genetics on early childhood bullying behavior. Participants were 561 children who participated in a longitudinal randomized control trial of a preventive intervention beginning in first grade who were present for the first grade peer nominations used to measure early childhood bullying and who provided genetic data during the age 19-21 year follow-up in the form of blood or saliva. Measures included a polygenic risk score (PRS) derived from a conduct disorder genome wide association study. Latent profile analysis identified three profiles of bullying behaviors during early childhood. Results suggest that the PRS was significantly associated with class membership, with individuals in the moderate bully-victim profile having the highest levels of the PRS and those in the high bully-victim profile having the lowest levels. This line of research has important implications for understanding genetic vulnerability to bullying in early childhood.
Collapse
Affiliation(s)
- Rashelle J Musci
- Department of Mental Health, Johns Hopkins University, Bloomberg School of Public Health, 624 North Broadway, Baltimore, MD 21205, USA.
| | - Amie F Bettencourt
- Department of Mental Health, Johns Hopkins University, Bloomberg School of Public Health, 624 North Broadway, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Division of Child and Adolescent Psychiatry, Johns Hopkins School of Medicine, 550 North Broadway, Baltimore, MD 21205, USA
| | - Danielle Sisto
- Department of Mental Health, Johns Hopkins University, Bloomberg School of Public Health, 624 North Broadway, Baltimore, MD 21205, USA
| | - Brion Maher
- Department of Mental Health, Johns Hopkins University, Bloomberg School of Public Health, 624 North Broadway, Baltimore, MD 21205, USA
| | - George Uhl
- Research Service, New Mexico VA Healthcare System, Departments of Neurology, Neuroscience and Molecular Genetics and Microbiology, University of New Mexico, Departments of Neurology, Neuroscience and Mental Health, Johns Hopkins Medical Institutions
| | - Nicholas Ialongo
- Department of Mental Health, Johns Hopkins University, Bloomberg School of Public Health, 624 North Broadway, Baltimore, MD 21205, USA
| | - Catherine P Bradshaw
- Department of Mental Health, Johns Hopkins University, Bloomberg School of Public Health, 624 North Broadway, Baltimore, MD 21205, USA; Curry School of Education, University of Virginia, PO Box 400270, Charlottesille, VA 22904, USA
| |
Collapse
|
31
|
Tielbeek JJ, Johansson A, Polderman TJC, Rautiainen MR, Jansen P, Taylor M, Tong X, Lu Q, Burt AS, Tiemeier H, Viding E, Plomin R, Martin NG, Heath AC, Madden PAF, Montgomery G, Beaver KM, Waldman I, Gelernter J, Kranzler HR, Farrer LA, Perry JRB, Munafò M, LoParo D, Paunio T, Tiihonen J, Mous SE, Pappa I, de Leeuw C, Watanabe K, Hammerschlag AR, Salvatore JE, Aliev F, Bigdeli TB, Dick D, Faraone SV, Popma A, Medland SE, Posthuma D. Genome-Wide Association Studies of a Broad Spectrum of Antisocial Behavior. JAMA Psychiatry 2017; 74:1242-1250. [PMID: 28979981 PMCID: PMC6309228 DOI: 10.1001/jamapsychiatry.2017.3069] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Importance Antisocial behavior (ASB) places a large burden on perpetrators, survivors, and society. Twin studies indicate that half of the variation in this trait is genetic. Specific causal genetic variants have, however, not been identified. Objectives To estimate the single-nucleotide polymorphism-based heritability of ASB; to identify novel genetic risk variants, genes, or biological pathways; to test for pleiotropic associations with other psychiatric traits; and to reevaluate the candidate gene era data through the Broad Antisocial Behavior Consortium. Design, Setting, and Participants Genome-wide association data from 5 large population-based cohorts and 3 target samples with genome-wide genotype and ASB data were used for meta-analysis from March 1, 2014, to May 1, 2016. All data sets used quantitative phenotypes, except for the Finnish Crime Study, which applied a case-control design (370 patients and 5850 control individuals). Main Outcome and Measures This study adopted relatively broad inclusion criteria to achieve a quantitative measure of ASB derived from multiple measures, maximizing the sample size over different age ranges. Results The discovery samples comprised 16 400 individuals, whereas the target samples consisted of 9381 individuals (all individuals were of European descent), including child and adult samples (mean age range, 6.7-56.1 years). Three promising loci with sex-discordant associations were found (8535 female individuals, chromosome 1: rs2764450, chromosome 11: rs11215217; 7772 male individuals, chromosome X, rs41456347). Polygenic risk score analyses showed prognostication of antisocial phenotypes in an independent Finnish Crime Study (2536 male individuals and 3684 female individuals) and shared genetic origin with conduct problems in a population-based sample (394 male individuals and 431 female individuals) but not with conduct disorder in a substance-dependent sample (950 male individuals and 1386 female individuals) (R2 = 0.0017 in the most optimal model, P = 0.03). Significant inverse genetic correlation of ASB with educational attainment (r = -0.52, P = .005) was detected. Conclusions and Relevance The Broad Antisocial Behavior Consortium entails the largest collaboration to date on the genetic architecture of ASB, and the first results suggest that ASB may be highly polygenic and has potential heterogeneous genetic effects across sex.
Collapse
Affiliation(s)
- Jorim J Tielbeek
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Child and Adolescent Psychiatry, VU University Medical Center, Amsterdam, the Netherlands
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ada Johansson
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Psychology, Faculty of Arts, Psychology, and Theology, Åbo Akademi University, Turku, Finland
| | - Tinca J C Polderman
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Marja-Riitta Rautiainen
- National Institute for Health and Welfare, Helsinki, Finland
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, Kuopio
- Department of Psychiatry, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Philip Jansen
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Michelle Taylor
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, England
| | - Xiaoran Tong
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing
| | - Qing Lu
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing
| | - Alexandra S Burt
- Department of Psychology, Michigan State University, East Lansing
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Essi Viding
- Division of Psychology and Language Sciences, University College London, London, England
| | - Robert Plomin
- Division of Psychology and Language Sciences, University College London, London, England
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Andrew C Heath
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Pamela A F Madden
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Grant Montgomery
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Kevin M Beaver
- College of Criminology and Criminal Justice, Florida State University, Tallahassee
- Center for Social and Humanities Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Irwin Waldman
- Psychology Department, Emory University, Atlanta, Georgia
| | - Joel Gelernter
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs (VA) Connecticut Healthcare Center, New Haven
| | - Henry R Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts
| | - John R B Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, England
| | - Marcus Munafò
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, England
| | - Devon LoParo
- Psychology Department, Emory University, Atlanta, Georgia
| | - Tiina Paunio
- National Institute for Health and Welfare, Helsinki, Finland
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, Kuopio
- Department of Psychiatry, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jari Tiihonen
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, Kuopio, Finland
| | - Sabine E Mous
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Irene Pappa
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Christiaan de Leeuw
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Kyoko Watanabe
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Anke R Hammerschlag
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jessica E Salvatore
- Department of Psychology and the Virginia Institute for Psychiatric and Behavioural Genetics, Virginia Commonwealth University, Richmond
| | - Fazil Aliev
- Department of African American Studies, Virginia Commonwealth University, Richmond
- Faculty of Business, Karabuk University, Karabuk, Turkey
| | - Tim B Bigdeli
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond
| | - Danielle Dick
- Department of Psychology, African American Studies, and Human & Molecular Genetics, Virginia Commonwealth University, Richmond
| | - Stephen V Faraone
- Department of Psychiatry and Behavioral Sciences, Psychiatric Genetic Epidemiology and Neurobiology Laboratory, SUNY Upstate Medical University, Syracuse, New York
- Department of Neuroscience and Physiology, Psychiatric Genetic Epidemiology and Neurobiology Laboratory, SUNY Upstate Medical University, Syracuse, New York
| | - Arne Popma
- Department of Child and Adolescent Psychiatry, VU University Medical Center, Amsterdam, the Netherlands
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Genetics, Neuroscience Campus Amsterdam, Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
32
|
Zeng Y, Navarro P, Shirali M, Howard DM, Adams MJ, Hall LS, Clarke TK, Thomson PA, Smith BH, Murray A, Padmanabhan S, Hayward C, Boutin T, MacIntyre DJ, Lewis CM, Wray NR, Mehta D, Penninx BW, Milaneschi Y, Baune BT, Air T, Hottenga JJ, Mbarek H, Castelao E, Pistis G, Schulze TG, Streit F, Forstner AJ, Byrne EM, Martin NG, Breen G, Müller-Myhsok B, Lucae S, Kloiber S, Domenici E, Deary IJ, Porteous DJ, Haley CS, McIntosh AM. Genome-wide Regional Heritability Mapping Identifies a Locus Within the TOX2 Gene Associated With Major Depressive Disorder. Biol Psychiatry 2017; 82:312-321. [PMID: 28153336 PMCID: PMC5553996 DOI: 10.1016/j.biopsych.2016.12.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/16/2016] [Accepted: 12/13/2016] [Indexed: 12/03/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is the second largest cause of global disease burden. It has an estimated heritability of 37%, but published genome-wide association studies have so far identified few risk loci. Haplotype-block-based regional heritability mapping (HRHM) estimates the localized genetic variance explained by common variants within haplotype blocks, integrating the effects of multiple variants, and may be more powerful for identifying MDD-associated genomic regions. METHODS We applied HRHM to Generation Scotland: The Scottish Family Health Study, a large family- and population-based Scottish cohort (N = 19,896). Single-single nucleotide polymorphism (SNP) and haplotype-based association tests were used to localize the association signal within the regions identified by HRHM. Functional prediction was used to investigate the effect of MDD-associated SNPs within the regions. RESULTS A haplotype block across a 24-kb region within the TOX2 gene reached genome-wide significance in HRHM. Single-SNP- and haplotype-based association tests demonstrated that five of nine genotyped SNPs and two haplotypes within this block were significantly associated with MDD. The expression of TOX2 and a brain-specific long noncoding RNA RP1-269M15.3 in frontal cortex and nucleus accumbens basal ganglia, respectively, were significantly regulated by MDD-associated SNPs within this region. Both the regional heritability and single-SNP associations within this block were replicated in the UK-Ireland group of the most recent release of the Psychiatric Genomics Consortium (PGC), the PGC2-MDD (Major Depression Dataset). The SNP association was also replicated in a depressive symptom sample that shares some individuals with the PGC2-MDD. CONCLUSIONS This study highlights the value of HRHM for MDD and provides an important target within TOX2 for further functional studies.
Collapse
Affiliation(s)
- Yanni Zeng
- Division of Psychiatry, University of Edinburgh, Edinburgh.
| | - Pau Navarro
- Medical Research Council Human Genetics Unit, University of Edinburgh, Edinburgh
| | - Masoud Shirali
- Medical Research Council Human Genetics Unit, University of Edinburgh, Edinburgh,Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh
| | | | - Mark J. Adams
- Division of Psychiatry, University of Edinburgh, Edinburgh
| | - Lynsey S. Hall
- Division of Psychiatry, University of Edinburgh, Edinburgh
| | | | - Pippa A. Thomson
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh,Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh
| | - Blair H. Smith
- Department of Psychology, University of Edinburgh, Edinburgh,Division of Population Health Sciences, University of Dundee, Dundee
| | - Alison Murray
- Division of Applied Health Sciences, University of Aberdeen, Aberdeen
| | - Sandosh Padmanabhan
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh,Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow
| | - Caroline Hayward
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh
| | - Thibaud Boutin
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh
| | | | - Cathryn M. Lewis
- MRC Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Naomi R. Wray
- Queensland Brain Institute, University of Queensland, St. Lucia, Queensland
| | - Divya Mehta
- Queensland Brain Institute, University of Queensland, St. Lucia, Queensland
| | | | - Yuri Milaneschi
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Bernhard T. Baune
- Discipline of Psychiatry, University of Adelaide, Adelaide, Australia
| | - Tracy Air
- Discipline of Psychiatry, University of Adelaide, Adelaide, Australia
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Hamdi Mbarek
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Enrique Castelao
- Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Giorgio Pistis
- Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Thomas G. Schulze
- Institute of Psychiatric Phenomics and Genomics, Ludwig-Maximilians-University, Munich Cluster for Systems Neurology, Munich,Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University, Göttingen,Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim
| | - Andreas J. Forstner
- Institute of Human Genetics, Life and Brain Center, University of Bonn, Bonn, Germany,Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Enda M. Byrne
- Queensland Brain Institute, University of Queensland, St. Lucia, Queensland
| | | | - Gerome Breen
- MRC Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | | | - Susanne Lucae
- Max Planck Institute of Psychiatry, Munich Cluster for Systems Neurology, Munich
| | - Stefan Kloiber
- Max Planck Institute of Psychiatry, Munich Cluster for Systems Neurology, Munich
| | - Enrico Domenici
- Laboratory of Neurogenomic Biomarkers, Centre for Integrative Biology, University of Trento, Trento, Italy
| | | | - Ian J. Deary
- Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh,Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh,Department of Psychology, University of Edinburgh, Edinburgh
| | - David J. Porteous
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh,Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh,Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh
| | - Chris S. Haley
- Medical Research Council Human Genetics Unit, University of Edinburgh, Edinburgh,The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh
| | - Andrew M. McIntosh
- Division of Psychiatry, University of Edinburgh, Edinburgh,Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh,Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh
| |
Collapse
|
33
|
Petersen PS, Lei X, Wolf RM, Rodriguez S, Tan SY, Little HC, Schweitzer MA, Magnuson TH, Steele KE, Wong GW. CTRP7 deletion attenuates obesity-linked glucose intolerance, adipose tissue inflammation, and hepatic stress. Am J Physiol Endocrinol Metab 2017; 312:E309-E325. [PMID: 28223291 PMCID: PMC5406989 DOI: 10.1152/ajpendo.00344.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/10/2017] [Accepted: 02/06/2017] [Indexed: 12/22/2022]
Abstract
Chronic low-grade inflammation and cellular stress are important contributors to obesity-linked metabolic dysfunction. Here, we uncover an immune-metabolic role for C1q/TNF-related protein 7 (CTRP7), a secretory protein of the C1q family with previously unknown function. In obese humans, circulating CTRP7 levels were markedly elevated and positively correlated with body mass index, glucose, insulin, insulin resistance index, hemoglobin A1c, and triglyceride levels. Expression of CTRP7 in liver was also significantly upregulated in obese humans and positively correlated with gluconeogenic genes. In mice, Ctrp7 expression was differentially modulated in various tissues by fasting and refeeding and by diet-induced obesity. A genetic loss-of-function mouse model was used to determine the requirement of CTRP7 for metabolic homeostasis. When fed a control low-fat diet, male or female mice lacking CTRP7 were indistinguishable from wild-type littermates. In obese male mice consuming a high-fat diet, however, CTRP7 deficiency attenuated insulin resistance and enhanced glucose tolerance, effects that were independent of body weight, metabolic rate, and physical activity level. Improved glucose metabolism in CTRP7-deficient mice was associated with reduced adipose tissue inflammation, as well as decreased liver fibrosis and cellular oxidative and endoplasmic reticulum stress. These results provide a link between elevated CTRP7 levels and impaired glucose metabolism, frequently associated with obesity. Inhibiting CTRP7 action may confer beneficial metabolic outcomes in the setting of obesity and diabetes.
Collapse
Affiliation(s)
- Pia S Petersen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xia Lei
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Risa M Wolf
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stefanie Y Tan
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hannah C Little
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael A Schweitzer
- Department of Surgery, Johns Hopkins Center for Bariatric Surgery, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Thomas H Magnuson
- Department of Surgery, Johns Hopkins Center for Bariatric Surgery, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Kimberley E Steele
- Department of Surgery, Johns Hopkins Center for Bariatric Surgery, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland;
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
34
|
Bagshaw ATM, Horwood LJ, Fergusson DM, Gemmell NJ, Kennedy MA. Microsatellite polymorphisms associated with human behavioural and psychological phenotypes including a gene-environment interaction. BMC MEDICAL GENETICS 2017; 18:12. [PMID: 28158988 PMCID: PMC5291968 DOI: 10.1186/s12881-017-0374-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/25/2017] [Indexed: 02/05/2023]
Abstract
Background The genetic and environmental influences on human personality and behaviour are a complex matter of ongoing debate. Accumulating evidence indicates that short tandem repeats (STRs) in regulatory regions are good candidates to explain heritability not accessed by genome-wide association studies. Methods We tested for associations between the genotypes of four selected repeats and 18 traits relating to personality, behaviour, cognitive ability and mental health in a well-studied longitudinal birth cohort (n = 458-589) using one way analysis of variance. The repeats were a highly conserved poly-AC microsatellite in the upstream promoter region of the T-box brain 1 (TBR1) gene and three previously studied STRs in the activating enhancer-binding protein 2-beta (AP2-β) and androgen receptor (AR) genes. Where significance was found we used multiple regression to assess the influence of confounding factors. Results Carriers of the shorter, most common, allele of the AR gene’s GGN microsatellite polymorphism had fewer anxiety-related symptoms, which was consistent with previous studies, but in our study this was not significant following Bonferroni correction. No associations with two repeats in the AP2-β gene withstood this correction. A novel finding was that carriers of the minor allele of the TBR1 AC microsatellite were at higher risk of conduct problems in childhood at age 7-9 (p = 0.0007, which did pass Bonferroni correction). Including maternal smoking during pregnancy (MSDP) in models controlling for potentially confounding influences showed that an interaction between TBR1 genotype and MSDP was a significant predictor of conduct problems in childhood and adolescence (p < 0.001), and of self-reported criminal behaviour up to age 25 years (p ≤ 0.02). This interaction remained significant after controlling for possible confounders including maternal age at birth, socio-economic status and education, and offspring birth weight. Conclusions The potential functional importance of the TBR1 gene’s promoter microsatellite deserves further investigation. Our results suggest that it participates in a gene-environment interaction with MDSP and antisocial behaviour. However, previous evidence that mothers who smoke during pregnancy carry genes for antisocial behaviour suggests that epistasis may influence the interaction. Electronic supplementary material The online version of this article (doi:10.1186/s12881-017-0374-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrew T M Bagshaw
- Department of Pathology, University of Otago, Christchurch, PO Box 4345, Christchurch, New Zealand.
| | - L John Horwood
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - David M Fergusson
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Neil J Gemmell
- Department of Anatomy, University of Otago, Dunedin, New Zealand.,Gravida - National Centre for Growth and Development, University of Otago, Dunedin, New Zealand
| | - Martin A Kennedy
- Department of Pathology, University of Otago, Christchurch, PO Box 4345, Christchurch, New Zealand
| |
Collapse
|
35
|
Mann FD, Engelhardt L, Briley DA, Grotzinger AD, Patterson MW, Tackett JL, Strathan DB, Heath A, Lynskey M, Slutske W, Martin NG, Tucker-Drob EM, Harden KP. Sensation seeking and impulsive traits as personality endophenotypes for antisocial behavior: Evidence from two independent samples. PERSONALITY AND INDIVIDUAL DIFFERENCES 2017; 105:30-39. [PMID: 28824215 PMCID: PMC5560504 DOI: 10.1016/j.paid.2016.09.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Sensation seeking and impulsivity are personality traits that are correlated with risk for antisocial behavior (ASB). This paper uses two independent samples of twins to (a) test the extent to which sensation seeking and impulsivity statistically mediate genetic influence on ASB, and (b) compare this to genetic influences accounted for by other personality traits. In Sample 1, delinquent behavior, as well as impulsivity, sensation seeking and Big Five personality traits, were measured in adolescent twins from the Texas Twin Project. In Sample 2, adult twins from the Australian Twin Registry responded to questionnaires that assessed individual differences in Eysenck's and Cloninger's personality dimensions, and a structured telephone interview that asked participants to retrospectively report DSM-defined symptoms of conduct disorder. Bivariate quantitative genetic models were used to identify genetic overlap between personality traits and ASB. Across both samples, novelty/sensation seeking and impulsive traits accounted for larger portions of genetic variance in ASB than other personality traits. We discuss whether sensation seeking and impulsive personality are causal endophenotypes for ASB, or merely index genetic liability for ASB.
Collapse
Affiliation(s)
- Frank D. Mann
- Department of Psychology, University of Texas at Austin, Austin, TX, United States
| | - Laura Engelhardt
- Department of Psychology, University of Texas at Austin, Austin, TX, United States
| | - Daniel A. Briley
- Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Andrew D. Grotzinger
- Department of Psychology, University of Texas at Austin, Austin, TX, United States
| | - Megan W. Patterson
- Department of Psychology, University of Texas at Austin, Austin, TX, United States
| | - Jennifer L. Tackett
- Department of Psychology, Northwestern University, Evanston, IL, United States
| | - Dixie B. Strathan
- Faculty of Arts and Business, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Andrew Heath
- Psychiatry, Washington University School of Medicine, St Louis, MI, United States
| | | | - Wendy Slutske
- Department of Psychological Sciences, University of Missouri, Columbia, MO, United States
| | - Nicholas G. Martin
- Genetic Epidemiology, Molecular Epidemiology and Neurogenetics Laboratories, Queensland Institute of Medial Research, Brisbane, Queensland, Australia
| | - Elliot M. Tucker-Drob
- Department of Psychology, University of Texas at Austin, Austin, TX, United States
- Population Research Center, University of Texas at Austin, Austin, TX, United States
| | - K. Paige Harden
- Department of Psychology, University of Texas at Austin, Austin, TX, United States
- Population Research Center, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
36
|
Dick DM, Adkins AE, Kuo SIC. Genetic influences on adolescent behavior. Neurosci Biobehav Rev 2016; 70:198-205. [PMID: 27422449 PMCID: PMC5074858 DOI: 10.1016/j.neubiorev.2016.07.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/30/2016] [Accepted: 07/06/2016] [Indexed: 11/13/2022]
Abstract
Adolescence is a transitional, developmental phase with marked shifts in behavior, particularly as related to risk-taking and experimentation. Genetic influences on adolescent behavior also show marked changes across this developmental period; in fact, adolescence showcases the dynamic nature of genetic influences on human behavior. Using the twin studies literature on alcohol use and misuse, we highlight several principles of genetic influence on adolescent behavior. We illustrate how genetic influences change (increase) across adolescence, as individuals have more freedom to express their predispositions and to shape their social worlds. We show how there are multiple genetic pathways to risk, and how the environment can moderate the importance of genetic predispositions. Finally, we review the literature aimed at identifying specific genes involved in adolescent behavior and understanding how identified genes impact adolescent outcomes. Ultimately, understanding how genetic predispositions combine with environmental influences to impact pathways of risk and resilience should be translated into improved prevention and intervention efforts; this remains a rich area for future research.
Collapse
Affiliation(s)
- Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, 806 W. Franklin Street, Richmond, VA 23284, United States; Department of African American Studies, Virginia Commonwealth University, 816 W. Franklin Street, Richmond, VA 23284, United States; Department of Human & Molecular Genetics, Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, VA 23298, United States; College Behavioral and Emotional Health Institute, Virginia Commonwealth University, 816 W. Franklin Street, Richmond, VA 23284, United States.
| | - Amy E Adkins
- Department of Psychology, Virginia Commonwealth University, 806 W. Franklin Street, Richmond, VA 23284, United States; College Behavioral and Emotional Health Institute, Virginia Commonwealth University, 816 W. Franklin Street, Richmond, VA 23284, United States
| | - Sally I-Chun Kuo
- Department of Psychology, Virginia Commonwealth University, 806 W. Franklin Street, Richmond, VA 23284, United States
| |
Collapse
|
37
|
Rautiainen MR, Paunio T, Repo-Tiihonen E, Virkkunen M, Ollila HM, Sulkava S, Jolanki O, Palotie A, Tiihonen J. Genome-wide association study of antisocial personality disorder. Transl Psychiatry 2016; 6:e883. [PMID: 27598967 PMCID: PMC5048197 DOI: 10.1038/tp.2016.155] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 06/20/2016] [Accepted: 07/16/2016] [Indexed: 01/05/2023] Open
Abstract
The pathophysiology of antisocial personality disorder (ASPD) remains unclear. Although the most consistent biological finding is reduced grey matter volume in the frontal cortex, about 50% of the total liability to developing ASPD has been attributed to genetic factors. The contributing genes remain largely unknown. Therefore, we sought to study the genetic background of ASPD. We conducted a genome-wide association study (GWAS) and a replication analysis of Finnish criminal offenders fulfilling DSM-IV criteria for ASPD (N=370, N=5850 for controls, GWAS; N=173, N=3766 for controls and replication sample). The GWAS resulted in suggestive associations of two clusters of single-nucleotide polymorphisms at 6p21.2 and at 6p21.32 at the human leukocyte antigen (HLA) region. Imputation of HLA alleles revealed an independent association with DRB1*01:01 (odds ratio (OR)=2.19 (1.53-3.14), P=1.9 × 10(-5)). Two polymorphisms at 6p21.2 LINC00951-LRFN2 gene region were replicated in a separate data set, and rs4714329 reached genome-wide significance (OR=1.59 (1.37-1.85), P=1.6 × 10(-9)) in the meta-analysis. The risk allele also associated with antisocial features in the general population conditioned for severe problems in childhood family (β=0.68, P=0.012). Functional analysis in brain tissue in open access GTEx and Braineac databases revealed eQTL associations of rs4714329 with LINC00951 and LRFN2 in cerebellum. In humans, LINC00951 and LRFN2 are both expressed in the brain, especially in the frontal cortex, which is intriguing considering the role of the frontal cortex in behavior and the neuroanatomical findings of reduced gray matter volume in ASPD. To our knowledge, this is the first study showing genome-wide significant and replicable findings on genetic variants associated with any personality disorder.
Collapse
Affiliation(s)
- M-R Rautiainen
- National Institute for Health and Welfare, Department of Health, Helsinki, Finland,Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, Kuopio, Finland,Department of Psychiatry, University of Helsinki, Helsinki, Finland,Department of Psychiatry, Helsinki University Hospital, Helsinki, Finland
| | - T Paunio
- National Institute for Health and Welfare, Department of Health, Helsinki, Finland,Department of Psychiatry, University of Helsinki, Helsinki, Finland,Department of Psychiatry, Helsinki University Hospital, Helsinki, Finland,Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland,National Institute for Health and Welfare, Department of Health, PO Box 30, Helsinki FI-00271, FinlandE-mail:
| | - E Repo-Tiihonen
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, Kuopio, Finland
| | - M Virkkunen
- Department of Psychiatry, University of Helsinki, Helsinki, Finland,Department of Psychiatry, Helsinki University Hospital, Helsinki, Finland
| | - H M Ollila
- National Institute for Health and Welfare, Department of Health, Helsinki, Finland,Stanford University Center for Sleep Sciences, Palo Alto, CA, USA
| | - S Sulkava
- National Institute for Health and Welfare, Department of Health, Helsinki, Finland,Department of Psychiatry, University of Helsinki, Helsinki, Finland,Department of Psychiatry, Helsinki University Hospital, Helsinki, Finland
| | - O Jolanki
- Stanford University Center for Sleep Sciences, Palo Alto, CA, USA
| | - A Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland,Wellcome Trust Sanger Institute, Hinxton, UK,Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA,Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - J Tiihonen
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, Kuopio, Finland,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden,Karolinska Institutet, Department of Clinical Neuroscience, Byggnad R5, Stockholm S-171 76, Sweden. E-mail:
| |
Collapse
|
38
|
A Test-Replicate Approach to Candidate Gene Research on Addiction and Externalizing Disorders: A Collaboration Across Five Longitudinal Studies. Behav Genet 2016; 46:608-626. [PMID: 27444553 DOI: 10.1007/s10519-016-9800-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/06/2016] [Indexed: 10/21/2022]
Abstract
This study presents results from a collaboration across five longitudinal studies seeking to test and replicate models of gene-environment interplay in the development of substance use and externalizing disorders (SUDs, EXT). We describe an overview of our conceptual models, plan for gene-environment interplay analyses, and present main effects results evaluating six candidate genes potentially relevant to SUDs and EXT (MAOA, 5-HTTLPR, COMT, DRD2, DAT1, and DRD4). All samples included rich longitudinal and phenotypic measurements from childhood/adolescence (ages 5-13) through early adulthood (ages 25-33); sample sizes ranged from 3487 in the test sample, to ~600-1000 in the replication samples. Phenotypes included lifetime symptom counts of SUDs (nicotine, alcohol and cannabis), adult antisocial behavior, and an aggregate externalizing disorder composite. Covariates included the first 10 ancestral principal components computed using all autosomal markers in subjects across the data sets, and age at the most recent assessment. Sex, ancestry, and exposure effects were thoroughly evaluated. After correcting for multiple testing, only one significant main effect was found in the test sample, but it was not replicated. Implications for subsequent gene-environment interplay analyses are discussed.
Collapse
|
39
|
Brevik EJ, van Donkelaar MMJ, Weber H, Sánchez‐Mora C, Jacob C, Rivero O, Kittel‐Schneider S, Garcia‐Martínez I, Aebi M, van Hulzen K, Cormand B, Ramos‐Quiroga JA, Lesch K, Reif A, Ribasés M, Franke B, Posserud M, Johansson S, Lundervold AJ, Haavik J, Zayats T. Genome-wide analyses of aggressiveness in attention-deficit hyperactivity disorder. Am J Med Genet B Neuropsychiatr Genet 2016; 171:733-47. [PMID: 27021288 PMCID: PMC5071721 DOI: 10.1002/ajmg.b.32434] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/09/2016] [Indexed: 12/03/2022]
Abstract
Aggressiveness is a behavioral trait that has the potential to be harmful to individuals and society. With an estimated heritability of about 40%, genetics is important in its development. We performed an exploratory genome-wide association (GWA) analysis of childhood aggressiveness in attention deficit hyperactivity disorder (ADHD) to gain insight into the underlying biological processes associated with this trait. Our primary sample consisted of 1,060 adult ADHD patients (aADHD). To further explore the genetic architecture of childhood aggressiveness, we performed enrichment analyses of suggestive genome-wide associations observed in aADHD among GWA signals of dimensions of oppositionality (defiant/vindictive and irritable dimensions) in childhood ADHD (cADHD). No single polymorphism reached genome-wide significance (P < 5.00E-08). The strongest signal in aADHD was observed at rs10826548, within a long noncoding RNA gene (beta = -1.66, standard error (SE) = 0.34, P = 1.07E-06), closely followed by rs35974940 in the neurotrimin gene (beta = 3.23, SE = 0.67, P = 1.26E-06). The top GWA SNPs observed in aADHD showed significant enrichment of signals from both the defiant/vindictive dimension (Fisher's P-value = 2.28E-06) and the irritable dimension in cADHD (Fisher's P-value = 0.0061). In sum, our results identify a number of biologically interesting markers possibly underlying childhood aggressiveness and provide targets for further genetic exploration of aggressiveness across psychiatric disorders. © 2016 The Authors. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Erlend J. Brevik
- Division of PsychiatryHaukeland University HospitalBergenNorway
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of BiomedicineUniversity of BergenBergenNorway
- Department of Biological and Medical PsychologyUniversity of BergenBergenNorway
| | - Marjolein M. J. van Donkelaar
- Department of Human GeneticsDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
| | - Heike Weber
- Department of Psychiatry, Psychosomatics and PsychotherapyUniversity of FrankfurtFrankfurtGermany
| | - Cristina Sánchez‐Mora
- Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR)Universitat Autònoma de BarcelonaBarcelonaSpain
- Department of PsychiatryHospital Universitari Vall d'HebronBarcelonaSpain
- Biomedical Network Research Centre on Mental Health (CIBERSAM)BarcelonaSpain
| | - Christian Jacob
- Department of Psychiatry and PsychotherapyKlinik NürtingenNürtingenGermany
| | - Olga Rivero
- Division of Molecular PsychiatryCenter of Mental Health, University of WürzburgWürzburgGermany
| | - Sarah Kittel‐Schneider
- Division of Molecular PsychiatryCenter of Mental Health, University of WürzburgWürzburgGermany
| | - Iris Garcia‐Martínez
- Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR)Universitat Autònoma de BarcelonaBarcelonaSpain
- Department of PsychiatryHospital Universitari Vall d'HebronBarcelonaSpain
| | - Marcel Aebi
- Department of Forensic PsychiatryChild and Youth Forensic Service, University Hospital of PsychiatryZurichSwitzerland
- Department of Child and Adolescent PsychiatryUniversity of ZurichZurichSwitzerland
| | - Kimm van Hulzen
- Department of Human GeneticsDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
| | - Bru Cormand
- Facultat de Biologia, Departament de GenèticaUniversitat de BarcelonaCataloniaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)BarcelonaSpain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB)CataloniaSpain
| | - Josep A. Ramos‐Quiroga
- Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR)Universitat Autònoma de BarcelonaBarcelonaSpain
- Department of PsychiatryHospital Universitari Vall d'HebronBarcelonaSpain
- Biomedical Network Research Centre on Mental Health (CIBERSAM)BarcelonaSpain
- Department of Psychiatry and Legal MedicineUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Klaus‐Peter Lesch
- Department of Psychiatry and PsychotherapyKlinik NürtingenNürtingenGermany
- Department of Translational NeuroscienceSchool for Mental Health and Neuroscience (MHeNS), Maastricht UniversityMaastrichtThe Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatics and PsychotherapyUniversity of FrankfurtFrankfurtGermany
| | - Marta Ribasés
- Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR)Universitat Autònoma de BarcelonaBarcelonaSpain
- Department of PsychiatryHospital Universitari Vall d'HebronBarcelonaSpain
- Biomedical Network Research Centre on Mental Health (CIBERSAM)BarcelonaSpain
| | - Barbara Franke
- Department of Human GeneticsDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
- Department of PsychiatryDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
| | - Maj‐Britt Posserud
- Division of PsychiatryHaukeland University HospitalBergenNorway
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of BiomedicineUniversity of BergenBergenNorway
| | - Stefan Johansson
- Center for Medical Genetics and Molecular MedicineHaukeland University HospitalBergenNorway
- Department of Clinical ScienceUniversity of BergenBergenNorway
| | - Astri J. Lundervold
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of BiomedicineUniversity of BergenBergenNorway
- Department of Biological and Medical PsychologyUniversity of BergenBergenNorway
| | - Jan Haavik
- Division of PsychiatryHaukeland University HospitalBergenNorway
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of BiomedicineUniversity of BergenBergenNorway
| | - Tetyana Zayats
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of BiomedicineUniversity of BergenBergenNorway
| | | |
Collapse
|
40
|
Aebi M, van Donkelaar MMJ, Poelmans G, Buitelaar JK, Sonuga‐Barke EJS, Stringaris A, consortium IMAGE, Faraone SV, Franke B, Steinhausen H, van Hulzen KJE. Gene-set and multivariate genome-wide association analysis of oppositional defiant behavior subtypes in attention-deficit/hyperactivity disorder. Am J Med Genet B Neuropsychiatr Genet 2016; 171:573-88. [PMID: 26184070 PMCID: PMC4715802 DOI: 10.1002/ajmg.b.32346] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 06/29/2015] [Indexed: 12/02/2022]
Abstract
Oppositional defiant disorder (ODD) is a frequent psychiatric disorder seen in children and adolescents with attention-deficit-hyperactivity disorder (ADHD). ODD is also a common antecedent to both affective disorders and aggressive behaviors. Although the heritability of ODD has been estimated to be around 0.60, there has been little research into the molecular genetics of ODD. The present study examined the association of irritable and defiant/vindictive dimensions and categorical subtypes of ODD (based on latent class analyses) with previously described specific polymorphisms (DRD4 exon3 VNTR, 5-HTTLPR, and seven OXTR SNPs) as well as with dopamine, serotonin, and oxytocin genes and pathways in a clinical sample of children and adolescents with ADHD. In addition, we performed a multivariate genome-wide association study (GWAS) of the aforementioned ODD dimensions and subtypes. Apart from adjusting the analyses for age and sex, we controlled for "parental ability to cope with disruptive behavior." None of the hypothesis-driven analyses revealed a significant association with ODD dimensions and subtypes. Inadequate parenting behavior was significantly associated with all ODD dimensions and subtypes, most strongly with defiant/vindictive behaviors. In addition, the GWAS did not result in genome-wide significant findings but bioinformatics and literature analyses revealed that the proteins encoded by 28 of the 53 top-ranked genes functionally interact in a molecular landscape centered around Beta-catenin signaling and involved in the regulation of neurite outgrowth. Our findings provide new insights into the molecular basis of ODD and inform future genetic studies of oppositional behavior. © 2015 The Authors. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marcel Aebi
- Department of Forensic Psychiatry, Child and Youth Forensic ServiceUniversity Hospital of PsychiatryZurichSwitzerland
- Department of Child and Adolescent PsychiatryUniversity of ZurichZurichSwitzerland
| | - Marjolein M. J. van Donkelaar
- Department of Human GeneticsRadboud University Medical Center, Donders Institute for Brain, Cognition and BehaviourNijmegenThe Netherlands
| | - Geert Poelmans
- Department of Human GeneticsRadboud University Medical Center, Donders Institute for Brain, Cognition and BehaviourNijmegenThe Netherlands
- Department of Cognitive NeuroscienceDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
- Department of Molecular Animal PhysiologyDonders Institute for Brain, Cognition and Behavior, Radboud Institute for Molecular Life Sciences, Radboud UniversityNijmegenThe Netherlands
| | - Jan K. Buitelaar
- Department of Cognitive NeuroscienceDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
| | - Edmund J. S. Sonuga‐Barke
- Developmental Brain‐Behaviour LaboratoryDepartment of PsychologyUniversity of SouthamptonSouthamptonUK
- Department of Experimental Clinical and Health PsychologyGhent UniversityGhentBelgium
| | | | - IMAGE consortium
- Department of Forensic Psychiatry, Child and Youth Forensic ServiceUniversity Hospital of PsychiatryZurichSwitzerland
| | - Stephen V. Faraone
- Department of PsychiatrySUNY Upstate Medical UniversitySyracuseNew York
- Departmentof Neuroscience and PhysiologySUNY Upstate Medical UniversitySyracuseNew York
- Department of BiomedicineK.G. Jebsen Centre for Psychiatric DisordersUniversity of BergenBergenNorway
| | - Barbara Franke
- Department of Human GeneticsRadboud University Medical Center, Donders Institute for Brain, Cognition and BehaviourNijmegenThe Netherlands
- Department of PsychiatryDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
| | - Hans‐Christoph Steinhausen
- Department of Child and Adolescent PsychiatryUniversity of ZurichZurichSwitzerland
- Department of Psychology, Clinical Psychology and EpidemiologyUniversity of BaselBaselSwitzerland
- Research Unit for Child and Adolescent Psychiatry, Psychiatric HospitalAalborg University HospitalAalborgDenmark
| | - Kimm J. E. van Hulzen
- Department of Human GeneticsRadboud University Medical Center, Donders Institute for Brain, Cognition and BehaviourNijmegenThe Netherlands
| |
Collapse
|
41
|
Ehlers CL, Gizer IR, Bizon C, Slutske W, Peng Q, Schork NJ, Wilhelmsen KC. Single nucleotide polymorphisms in the REG-CTNNA2 region of chromosome 2 and NEIL3 associated with impulsivity in a Native American sample. GENES BRAIN AND BEHAVIOR 2016; 15:568-77. [PMID: 27167163 DOI: 10.1111/gbb.12297] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/18/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022]
Abstract
Impulsivity is a multi-faceted construct that, while characterized by a set of correlated dimensions, is centered around a core definition that involves acting suddenly in an unplanned manner without consideration for the consequences of such behavior. Several psychiatric disorders include impulsivity as a criterion, and thus it has been suggested that it may link a number of different behavioral disorders, including substance abuse. Native Americans (NA) experience some of the highest rates of substance abuse of all the US ethnic groups. The described analyses used data from a low-coverage whole genome sequence scan to conduct a genome-wide association study (GWAS) of an impulsivity phenotype in an American Indian community sample (n = 658). Demographic and clinical information were obtained using a semi-structured interview. Impulsivity was assessed using a scale derived from the Maudsley personality inventory that combines both novelty seeking and lack of planning items. The impulsivity score was tested for association with each variant adjusted for demographic variables, and corrected for ancestry and kinship, using emmax. Simulations were conducted to calculate empirical P-values. Genome-wide significant findings were observed for a variant 50-kb upstream from catenin cadherin-associated protein, alpha 2 (CTNNA2), a neuronal-specific catenin, in the REG gene cluster. A meta-analysis of GWAS had previously identified common variants in CTNNA2 as being associated with excitement seeking. A second locus upstream of nei endonuclease VIII-like 3 (NEIL3) on chromosome 4 also achieved genome-wide significance. The association between sequence variants in these regions suggests their potential roles in the genetic regulation of this phenotype in this population.
Collapse
Affiliation(s)
- C L Ehlers
- The Scripps Research Institute, Molecular and Cellular Neuroscience Department, La Jolla, CA
| | - I R Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO
| | - C Bizon
- Renaissance Computing Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - W Slutske
- Department of Psychological Sciences, University of Missouri, Columbia, MO
| | - Q Peng
- The Scripps Research Institute, Molecular and Cellular Neuroscience Department, La Jolla, CA.,J Craig Venter Institute, Human Biology, La Jolla, CA
| | - N J Schork
- J Craig Venter Institute, Human Biology, La Jolla, CA
| | - K C Wilhelmsen
- Renaissance Computing Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC.,Department of Genetics and Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
42
|
Few LR, Agrawal A. Commentary on Verweij et al. (2016): Conduct problems and substance use-genetic and environmental perspectives on sex differences. Addiction 2016; 111:1046-7. [PMID: 27157903 PMCID: PMC4941623 DOI: 10.1111/add.13371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 02/19/2016] [Indexed: 11/28/2022]
Abstract
Genes influence the covariance between conduct disorder (CD) problems and substance use in males, but shared environment is more important in females. We now need to consider: (a) how genetic influences on CD affect correlations across substances from early to later stages of involvement; (b) whether polygenic liability to CD and substance use overlap; (c) environmental pathways of vulnerability (e.g. peers) in females; and (d) sex effects derived from opposite-sex twin pairs.
Collapse
Affiliation(s)
- Lauren R. Few
- Washington University School of Medicine, Department of Psychiatry, 660 S. Euclid, CB 8134, Saint Louis, MO 63110
| | - Arpana Agrawal
- Washington University School of Medicine, Department of Psychiatry, St Louis, MO, USA.
| |
Collapse
|
43
|
Long Q, Argmann C, Houten SM, Huang T, Peng S, Zhao Y, Tu Z, Zhu J. Inter-tissue coexpression network analysis reveals DPP4 as an important gene in heart to blood communication. Genome Med 2016; 8:15. [PMID: 26856537 PMCID: PMC4746932 DOI: 10.1186/s13073-016-0268-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 01/21/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Inter-tissue molecular interactions are critical to the function and behavior of biological systems in multicellular organisms, but systematic studies of interactions between tissues are lacking. Also, existing studies of inter-tissue interactions are based on direct gene expression correlations, which can't distinguish correlations due to common genetic architectures versus biochemical or molecular signal exchange between tissues. METHODS We developed a novel strategy to study inter-tissue interaction by removing effects of genetic regulation of gene expression (genetic decorrelation). We applied our method to the comprehensive atlas of gene expression across nine human tissues in the Genotype-Tissue Expression (GTEx) project to generate novel genetically decorrelated inter-tissue networks. From this we derived modules of genes important in inter-tissue interactions that are likely driven by biological signal exchange instead of their common genetic basis. Importantly we highlighted communication between tissues and elucidated gene activities in one tissue inducing gene expression changes in others. RESULTS We reveal global unidirectional inter-tissue coordination of specific biological pathways such as protein synthesis. Using our data, we highlighted a clinically relevant example whereby heart expression of DPP4 was coordinated with a gene expression signature characteristic for whole blood proliferation, potentially impacting peripheral stem cell mobilization. We also showed that expression of the poorly characterized FOCAD in heart correlated with protein biosynthetic processes in the lung. CONCLUSIONS In summary, this is the first resource of human multi-tissue networks enabling the investigation of molecular inter-tissue interactions. With the networks in hand, we may systematically design combination therapies that simultaneously target multiple tissues or pinpoint potential side effects of a drug in other tissues.
Collapse
Affiliation(s)
- Quan Long
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sander M Houten
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tao Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Siwu Peng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yong Zhao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Zhidong Tu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
44
|
Weeland J, Overbeek G, de Castro BO, Matthys W. Underlying Mechanisms of Gene-Environment Interactions in Externalizing Behavior: A Systematic Review and Search for Theoretical Mechanisms. Clin Child Fam Psychol Rev 2015; 18:413-42. [PMID: 26537239 PMCID: PMC4637001 DOI: 10.1007/s10567-015-0196-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the last decade, several candidate genes (i.e., MAOA, DRD4, DRD2, DAT1, 5-HTTLPR, and COMT) have been extensively studied as potential moderators of the detrimental effects of postnatal family adversity on child externalizing behaviors, such as aggression and conduct disorder. Many studies on such candidate gene by environment interactions (i.e., cG × E) have been published, and the first part of this paper offers a systematic review and integration of their findings (n = 53). The overview shows a set of heterogeneous findings. However, because of large differences between studies in terms of sample composition, conceptualizations, and power, it is difficult to determine if different findings indeed illustrate inconsistent cG × E findings or if findings are simply incomparable. In the second part of the paper, therefore, we argue that one way to help resolve this problem is the development of theory-driven a priori hypotheses on which biopsychosocial mechanisms might underlie cG × E. Such a theoretically based approach can help us specify our research strategies, create more comparable findings, and help us interpret different findings between studies. In accordance, we describe three possible explanatory mechanisms, based on extant literature on the concepts of (1) emotional reactivity, (2) reward sensitivity, and (3) punishment sensitivity. For each mechanism, we discuss the link between the putative mechanism and externalizing behaviors, the genetic polymorphism, and family adversity. Possible research strategies to test these mechanisms, and implications for interventions, are discussed.
Collapse
Affiliation(s)
- Joyce Weeland
- Utrecht Centre for Child and Adolescent Studies, Utrecht University, PO Box 15.804, 1001 NH, Amsterdam, The Netherlands.
- Research Institute of Child Development and Education, University of Amsterdam, Amsterdam, The Netherlands.
| | - Geertjan Overbeek
- Research Institute of Child Development and Education, University of Amsterdam, Amsterdam, The Netherlands
| | - Bram Orobio de Castro
- Utrecht Centre for Child and Adolescent Studies, Utrecht University, PO Box 15.804, 1001 NH, Amsterdam, The Netherlands
| | - Walter Matthys
- Department of Child and Adolescent Studies, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
45
|
Abstract
Large-scale genomic investigations have just begun to illuminate the molecular genetic contributions to major psychiatric illnesses, ranging from small-effect-size common variants to larger-effect-size rare mutations. The findings provide causal anchors from which to understand their neurobiological basis. Although these studies represent enormous success, they highlight major challenges reflected in the heterogeneity and polygenicity of all of these conditions and the difficulty of connecting multiple levels of molecular, cellular, and circuit functions to complex human behavior. Nevertheless, these advances place us on the threshold of a new frontier in the pathophysiological understanding, diagnosis, and treatment of psychiatric disease.
Collapse
Affiliation(s)
- Daniel H Geschwind
- Departments of Neurology, Psychiatry, and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Jonathan Flint
- Wellcome Trust Center for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
46
|
Derringer J, Corley RP, Haberstick BC, Young SE, Demmitt BA, Howrigan DP, Kirkpatrick RM, Iacono WG, McGue M, Keller MC, Brown S, Tapert S, Hopfer CJ, Stallings MC, Crowley TJ, Rhee SH, Krauter K, Hewitt JK, McQueen MB. Genome-Wide Association Study of Behavioral Disinhibition in a Selected Adolescent Sample. Behav Genet 2015; 45:375-81. [PMID: 25637581 PMCID: PMC4459903 DOI: 10.1007/s10519-015-9705-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 01/07/2015] [Indexed: 10/24/2022]
Abstract
Behavioral disinhibition (BD) is a quantitative measure designed to capture the heritable variation encompassing risky and impulsive behaviors. As a result, BD represents an ideal target for discovering genetic loci that predispose individuals to a wide range of antisocial behaviors and substance misuse that together represent a large cost to society as a whole. Published genome-wide association studies (GWAS) have examined specific phenotypes that fall under the umbrella of BD (e.g. alcohol dependence, conduct disorder); however no GWAS has specifically examined the overall BD construct. We conducted a GWAS of BD using a sample of 1,901 adolescents over-selected for characteristics that define high BD, such as substance and antisocial behavior problems, finding no individual locus that surpassed genome-wide significance. Although no single SNP was significantly associated with BD, restricted maximum likelihood analysis estimated that 49.3 % of the variance in BD within the Caucasian sub-sample was accounted for by the genotyped SNPs (p = 0.06). Gene-based tests identified seven genes associated with BD (p ≤ 2.0 × 10(-6)). Although the current study was unable to identify specific SNPs or pathways with replicable effects on BD, the substantial sample variance that could be explained by all genotyped SNPs suggests that larger studies could successfully identify common variants associated with BD.
Collapse
Affiliation(s)
- Jaime Derringer
- Department of Psychology, University of Illinois Urbana-Champaign, Champaign, IL, 61820, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Jiang P, Scarpa JR, Fitzpatrick K, Losic B, Gao VD, Hao K, Summa KC, Yang HS, Zhang B, Allada R, Vitaterna MH, Turek FW, Kasarskis A. A systems approach identifies networks and genes linking sleep and stress: implications for neuropsychiatric disorders. Cell Rep 2015; 11:835-48. [PMID: 25921536 DOI: 10.1016/j.celrep.2015.04.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/23/2014] [Accepted: 03/30/2015] [Indexed: 02/06/2023] Open
Abstract
Sleep dysfunction and stress susceptibility are comorbid complex traits that often precede and predispose patients to a variety of neuropsychiatric diseases. Here, we demonstrate multilevel organizations of genetic landscape, candidate genes, and molecular networks associated with 328 stress and sleep traits in a chronically stressed population of 338 (C57BL/6J × A/J) F2 mice. We constructed striatal gene co-expression networks, revealing functionally and cell-type-specific gene co-regulations important for stress and sleep. Using a composite ranking system, we identified network modules most relevant for 15 independent phenotypic categories, highlighting a mitochondria/synaptic module that links sleep and stress. The key network regulators of this module are overrepresented with genes implicated in neuropsychiatric diseases. Our work suggests that the interplay among sleep, stress, and neuropathology emerges from genetic influences on gene expression and their collective organization through complex molecular networks, providing a framework for interrogating the mechanisms underlying sleep, stress susceptibility, and related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Peng Jiang
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Joseph R Scarpa
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Karrie Fitzpatrick
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Bojan Losic
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vance D Gao
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Keith C Summa
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - He S Yang
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ravi Allada
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Martha H Vitaterna
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Fred W Turek
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA.
| | - Andrew Kasarskis
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
48
|
Ye Z, Mayer J, Ivacic L, Zhou Z, He M, Schrodi SJ, Page D, Brilliant MH, Hebbring SJ. Phenome-wide association studies (PheWASs) for functional variants. Eur J Hum Genet 2014; 23:523-9. [PMID: 25074467 DOI: 10.1038/ejhg.2014.123] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 05/27/2014] [Accepted: 05/30/2014] [Indexed: 01/08/2023] Open
Abstract
The genome-wide association study (GWAS) is a powerful approach for studying the genetic complexities of human disease. Unfortunately, GWASs often fail to identify clinically significant associations and describing function can be a challenge. GWAS is a phenotype-to-genotype approach. It is now possible to conduct a converse genotype-to-phenotype approach using extensive electronic medical records to define a phenome. This approach associates a single genetic variant with many phenotypes across the phenome and is called a phenome-wide association study (PheWAS). The majority of PheWASs conducted have focused on variants identified previously by GWASs. This approach has been efficient for rediscovering gene-disease associations while also identifying pleiotropic effects for some single-nucleotide polymorphisms (SNPs). However, the use of SNPs identified by GWAS in a PheWAS is limited by the inherent properties of the GWAS SNPs, including weak effect sizes and difficulty when translating discoveries to function. To address these challenges, we conducted a PheWAS on 105 presumed functional stop-gain and stop-loss variants genotyped on 4235 Marshfield Clinic patients. Associations were validated on an additional 10 640 Marshfield Clinic patients. PheWAS results indicate that a nonsense variant in ARMS2 (rs2736911) is associated with age-related macular degeneration (AMD). These results demonstrate that focusing on functional variants may be an effective approach when conducting a PheWAS.
Collapse
Affiliation(s)
- Zhan Ye
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, WI, USA
| | - John Mayer
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, WI, USA
| | - Lynn Ivacic
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI, USA
| | - Zhiyi Zhou
- Parkland Center for Clinical Innovation, Parkland Health and Hospital System, Dallas, TX, USA
| | - Min He
- 1] Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, WI, USA [2] Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI, USA
| | - Steven J Schrodi
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI, USA
| | - David Page
- Computation and Informatics in Biology and Medicine, University of Wisconsin Madison, Madison, WI, USA
| | - Murray H Brilliant
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI, USA
| | - Scott J Hebbring
- 1] Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI, USA [2] Computation and Informatics in Biology and Medicine, University of Wisconsin Madison, Madison, WI, USA
| |
Collapse
|
49
|
Vrieze SI, Feng S, Miller MB, Hicks BM, Pankratz N, Abecasis GR, Iacono WG, McGue M. Rare nonsynonymous exonic variants in addiction and behavioral disinhibition. Biol Psychiatry 2014; 75:783-9. [PMID: 24094508 PMCID: PMC3975816 DOI: 10.1016/j.biopsych.2013.08.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/02/2013] [Accepted: 08/26/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Substance use is heritable, but few common genetic variants have been associated with these behaviors. Rare nonsynonymous exonic variants can now be efficiently genotyped, allowing exome-wide association tests. We identified and tested 111,592 nonsynonymous exonic variants for association with behavioral disinhibition and the use/misuse of nicotine, alcohol, and illicit drugs. METHODS Comprehensive genotyping of exonic variation combined with single-variant and gene-based tests of association was conducted in 7181 individuals; 172 candidate addiction genes were evaluated in greater detail. We also evaluated the aggregate effects of nonsynonymous variants on these phenotypes using Genome-wide Complex Trait Analysis. RESULTS No variant or gene was significantly associated with any phenotype. No association was found for any of the 172 candidate genes, even at reduced significance thresholds. All nonsynonymous variants jointly accounted for 35% of the heritability in illicit drug use and, when combined with common variants from a genome-wide array, accounted for 84% of the heritability. CONCLUSIONS Rare nonsynonymous variants may be important in etiology of illicit drug use, but detection of individual variants will require very large samples.
Collapse
Affiliation(s)
- Scott I Vrieze
- Center for Statistical Genetics (SIV, SF, GRA), Department of Biostatistics, University of Michigan, Ann Arbor, Michigan.
| | - Shuang Feng
- Center for Statistical Genetics (SIV, SF, GRA), Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Michael B Miller
- Department of Psychology (MBM, WGI, MM), University of Minnesota, Minneapolis, Minnesota
| | - Brian M Hicks
- Department of Psychiatry (BMH), University of Michigan, Ann Arbor, Michigan
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology (NP), University of Minnesota, Minneapolis, Minnesota
| | - Gonçalo R Abecasis
- Center for Statistical Genetics (SIV, SF, GRA), Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - William G Iacono
- Department of Psychology (MBM, WGI, MM), University of Minnesota, Minneapolis, Minnesota
| | - Matt McGue
- Department of Psychology (MBM, WGI, MM), University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
50
|
Systematic meta-analyses and field synopsis of genetic association studies of violence and aggression. Mol Psychiatry 2014; 19:471-7. [PMID: 23546171 PMCID: PMC3965568 DOI: 10.1038/mp.2013.31] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 01/21/2013] [Accepted: 02/01/2013] [Indexed: 12/15/2022]
Abstract
A large number of candidate gene studies for aggression and violence have been conducted. Successful identification of associations between genetic markers and aggression would contribute to understanding the neurobiology of antisocial behavior and potentially provide useful tools for risk prediction and therapeutic targets for high-risk groups of patients and offenders. We systematically reviewed the literature and assessed the evidence on genetic association studies of aggression and related outcomes in order to provide a field synopsis. We searched PubMed and Huge Navigator databases and sought additional data through reviewing reference lists and correspondence with investigators. Genetic association studies were included if outcome data on aggression or violent behavior either as a binary outcome or as a quantitative trait were provided. From 1331 potentially relevant investigations, 185 studies constituting 277 independent associations on 31 genes fulfilled the predetermined selection criteria. Data from variants investigated in three or more samples were combined in meta-analyses and potential sources of heterogeneity were investigated using subgroup analyses. In the primary analyses, which used relaxed inclusion criteria, we found no association between any polymorphism analyzed and aggression at the 5% level of significance. Subgroup analyses, including by severity of outcome, age group, characteristics of the sample and ethnicity, did not demonstrate any consistent findings. Current evidence does not support the use of such genes to predict dangerousness or as markers for therapeutic interventions.
Collapse
|