1
|
Wu CS, Tsai TH, Chen WL, Tsai HJ, Chien YL. Ophthalmologic diagnoses in youths with autism spectrum disorder: Prevalence and clinical correlates. Autism Res 2023; 16:2008-2020. [PMID: 37632715 DOI: 10.1002/aur.3019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/10/2023] [Indexed: 08/28/2023]
Abstract
Autism spectrum disorder (ASD) is associated with a high prevalence of visual dysfunction. This study aimed to investigate the rates of amblyopia, refractive errors, and strabismus, as well as their clinical correlates in ASD. This population-based matched-cohort study used data from the Taiwan National Health Insurance Research Database. A total of 3,551 youths with ASD and 35,510 non-autistic control participants matched by age and sex were included. All the participants were followed-up until they were 18 years old. The prevalence of amblyopia, refractive errors, and strabismus was compared between the ASD and control groups. Effect modifiers, including sex, ASD subgroup, and co-diagnosis of intelligence disability, were examined. Compared to the control group, youths with ASD had a significantly increased risk of amblyopia (adjusted odds ratio [aOR] = 1.75), anisometropia (aOR = 1.66), astigmatism (aOR = 1.51), hypermetropia (aOR = 2.08), exotropia (aOR = 2.86), and esotropia (aOR = 2.63), but a comparable likelihood of myopia according to age. Males with ASD had a significantly lower likelihood of exotropia, but a higher likelihood of myopia than females with ASD. The autism subgroup had a higher OR for hypermetropia, but a lower OR for myopia than the other ASD subgroups. ASD youths with intelligence disabilities demonstrated significantly higher ORs for amblyopia, hypermetropia, and all types of strabismus and lower OR for myopia than those without intelligence disabilities. In conclusion, the rates of amblyopia, refractive errors, and strabismus were higher in youths with ASD. Ocular abnormalities in youths with ASD require a comprehensive assessment and management.
Collapse
Affiliation(s)
- Chi-Shin Wu
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin, Taiwan
- Department of Psychiatry, National Taiwan University Hospital, Yunlin Branch, Yunlin, Taiwan
| | - Tzu-Hsun Tsai
- Department of Ophthalmology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Wei-Li Chen
- Department of Ophthalmology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- Advanced Ocular Surface and Corneal Nerve Regeneration Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Hui-Ju Tsai
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Yi-Ling Chien
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| |
Collapse
|
2
|
Lawther AJ, Zieba J, Fang Z, Furlong TM, Conn I, Govindaraju H, Choong LLY, Turner N, Siddiqui KS, Bridge W, Merlin S, Hyams TC, Killingsworth M, Eapen V, Clarke RA, Walker AK. Antioxidant Behavioural Phenotype in the Immp2l Gene Knock-Out Mouse. Genes (Basel) 2023; 14:1717. [PMID: 37761857 PMCID: PMC10531238 DOI: 10.3390/genes14091717] [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: 07/18/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Mitochondrial dysfunction is strongly associated with autism spectrum disorder (ASD) and the Inner mitochondrial membrane protein 2-like (IMMP2L) gene is linked to autism inheritance. However, the biological basis of this linkage is unknown notwithstanding independent reports of oxidative stress in association with both IMMP2L and ASD. To better understand IMMP2L's association with behaviour, we developed the Immp2lKD knockout (KO) mouse model which is devoid of Immp2l peptidase activity. Immp2lKD -/- KO mice do not display any of the core behavioural symptoms of ASD, albeit homozygous Immp2lKD -/- KO mice do display increased auditory stimulus-driven instrumental behaviour and increased amphetamine-induced locomotion. Due to reports of increased ROS and oxidative stress phenotypes in an earlier truncated Immp2l mouse model resulting from an intragenic deletion within Immp2l, we tested whether high doses of the synthetic mitochondrial targeted antioxidant (MitoQ) could reverse or moderate the behavioural changes in Immp2lKD -/- KO mice. To our surprise, we observed that ROS levels were not increased but significantly lowered in our new Immp2lKD -/- KO mice and that these mice had no oxidative stress-associated phenotypes and were fully fertile with no age-related ataxia or neurodegeneration as ascertained using electron microscopy. Furthermore, the antioxidant MitoQ had no effect on the increased amphetamine-induced locomotion of these mice. Together, these findings indicate that the behavioural changes in Immp2lKD -/- KO mice are associated with an antioxidant-like phenotype with lowered and not increased levels of ROS and no oxidative stress-related phenotypes. This suggested that treatments with antioxidants are unlikely to be effective in treating behaviours directly resulting from the loss of Immp2l/IMMP2L activity, while any behavioural deficits that maybe associated with IMMP2L intragenic deletion-associated truncations have yet to be determined.
Collapse
Affiliation(s)
- Adam J. Lawther
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Jerzy Zieba
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, NSW 2031, Australia
- Department of Psychology, University of Rzeszow, 35-310 Rzeszow, Poland
| | - Zhiming Fang
- Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW 2052, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW 2170, Australia; (T.C.H.)
| | - Teri M. Furlong
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Illya Conn
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, NSW 2031, Australia
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Hemna Govindaraju
- Department of Pharmacology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Laura L. Y. Choong
- Department of Pharmacology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Nigel Turner
- Department of Pharmacology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Khawar Sohail Siddiqui
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Wallace Bridge
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Sam Merlin
- Medical Science, School of Science, Western Sydney University, Campbelltown, Sydney, NSW 2751, Australia
| | - Tzipi Cohen Hyams
- Ingham Institute for Applied Medical Research, Sydney, NSW 2170, Australia; (T.C.H.)
| | - Murray Killingsworth
- Ingham Institute for Applied Medical Research, Sydney, NSW 2170, Australia; (T.C.H.)
- NSW Health Pathology, Liverpool Hospital Campus, 1 Campbell Street, Liverpool, NSW 2107, Australia
| | - Valsamma Eapen
- Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW 2052, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW 2170, Australia; (T.C.H.)
- Academic Unit of Infant Child and Adolescent Services (AUCS), South Western Sydney Local Health District, Liverpool, NSW 2170, Australia
| | - Raymond A. Clarke
- Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW 2052, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW 2170, Australia; (T.C.H.)
- Academic Unit of Infant Child and Adolescent Services (AUCS), South Western Sydney Local Health District, Liverpool, NSW 2170, Australia
| | - Adam K. Walker
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, NSW 2031, Australia
- Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW 2052, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
3
|
Mpoulimari I, Zintzaras E. Synthesis of genetic association studies on autism spectrum disorders using a genetic model-free approach. Psychiatr Genet 2022; 32:91-104. [PMID: 35353796 DOI: 10.1097/ypg.0000000000000316] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a clinically and genetically heterogeneous group of neurodevelopmental disorders. Despite the extensive efforts of scientists, the etiology of ASD is far from completely elucidated. In an effort to enlighten the genetic architecture of ASDs, a meta-analysis of all available genetic association studies (GAS) was conducted. METHODS We searched in the Human Genome Epidemiology Navigator (HuGE Navigator) and PubMed for available case-control GAS of ASDs. The threshold for meta-analysis was two studies per genetic variant. The association between genotype distribution and ASDs was examined using the generalized linear odds ratio (ORG). For variants with available allele frequencies, the examined model was the allele contrast. RESULTS Overall, 57 candidate genes and 128 polymorphisms were investigated in 159 articles. In total 28 genetic polymorphisms have been shown to be associated with ASDs, that are harbored in 19 genes. Statistically significant results were revealed for the variants of the following genes adenosine deaminase (ADA), bone marrow stromal cell antigen-1 (CD157/BST1), Dopamine receptor D1 (DRD1), engrailed homolog 2 (EN2), met proto-oncogene (MET), methylenetetrahydrofolate reductase (MTHFR), solute carrier family 6 member 4 (SLC6A4), Synaptosomal-associated protein, 25kDa (SNAP-25) and vitamin D receptor (VDR). In the allele contrast model of cases versus healthy controls, significant associations were observed for Adrenoceptor Alpha 1B (ADRA1B), acetyl serotonin O - methyltransferase (ASMT), complement component 4B (C4B), dopamine receptor D3 (DRD3), met proto-oncogene (MET), neuroligin 4, X-linked (NLGN4), neurexin 1 (NRXN1), oxytocin receptor (OXTR), Serine/Threonine-Protein Kinase PFTAIRE-1 (PFTK1), Reelin (RELN) and Ras-like without CAAX 2 (RIT2). CONCLUSION These significant findings provide further evidence for genetic factors' implication in ASDs offering new perspectives in means of prevention and prognosis.
Collapse
Affiliation(s)
- Ioanna Mpoulimari
- Department of Biomathematics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Elias Zintzaras
- Department of Biomathematics, Faculty of Medicine, University of Thessaly, Larissa, Greece
- Department of Medicine, The Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Super-conserved receptors expressed in the brain: biology and medicinal chemistry efforts. Future Med Chem 2022; 14:899-913. [PMID: 35535715 DOI: 10.4155/fmc-2022-0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The super-conserved receptors expressed in the brain (SREB) constitute a family of orphan G protein-coupled receptors that include GPR27 (SREB1), GPR85 (SREB2) and GPR173 (SREB3). Their sequences are highly conserved in vertebrates, and they are almost exclusively expressed in the central nervous system. This family of receptors has attracted much attention due to their putative physiological functions and their potential as novel drug targets. The SREB family has been postulated to play important roles in a wide range of different diseases, including pancreatic β-cell insulin secretion and regulation, schizophrenia, autism and atherosclerosis. This review intends to provide a comprehensive overview of the SREB family and its recent advances in biology and medicinal chemistry.
Collapse
|
5
|
Despang P, Salamon S, Breitenkamp A, Kuzmenkina E, Matthes J. Inhibitory effects on L- and N-type calcium channels by a novel Ca Vβ 1 variant identified in a patient with autism spectrum disorder. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:459-470. [PMID: 35122502 PMCID: PMC8873119 DOI: 10.1007/s00210-022-02213-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 01/25/2022] [Indexed: 12/01/2022]
Abstract
Voltage-gated calcium channel (VGCC) subunits have been genetically associated with autism spectrum disorders (ASD). The properties of the pore-forming VGCC subunit are modulated by auxiliary β-subunits, which exist in four isoforms (CaVβ1-4). Our previous findings suggested that activation of L-type VGCCs is a common feature of CaVβ2 subunit mutations found in ASD patients. In the current study, we functionally characterized a novel CaVβ1b variant (p.R296C) identified in an ASD patient. We used whole-cell and single-channel patch clamp to study the effect of CaVβ1b_R296C on the function of L- and N-type VGCCs. Furthermore, we used co-immunoprecipitation followed by Western blot to evaluate the interaction of the CaVβ1b-subunits with the RGK-protein Gem. Our data obtained at both, whole-cell and single-channel levels, show that compared to a wild-type CaVβ1b, the CaVβ1b_R296C variant inhibits L- and N-type VGCCs. Interaction with and modulation by the RGK-protein Gem seems to be intact. Our findings indicate functional effects of the CaVβ1b_R296C variant differing from that attributed to CaVβ2 variants found in ASD patients. Further studies have to detail the effects on different VGCC subtypes and on VGCC expression.
Collapse
Affiliation(s)
- Patrick Despang
- Center of Pharmacology, Institute II, University of Cologne, Gleueler Strasse 24, 50931, Köln, Cologne, Germany
| | - Sarah Salamon
- Center of Pharmacology, Institute II, University of Cologne, Gleueler Strasse 24, 50931, Köln, Cologne, Germany
| | - Alexandra Breitenkamp
- Center of Pharmacology, Institute II, University of Cologne, Gleueler Strasse 24, 50931, Köln, Cologne, Germany
| | - Elza Kuzmenkina
- Center of Pharmacology, Institute II, University of Cologne, Gleueler Strasse 24, 50931, Köln, Cologne, Germany
| | - Jan Matthes
- Center of Pharmacology, Institute II, University of Cologne, Gleueler Strasse 24, 50931, Köln, Cologne, Germany.
| |
Collapse
|
6
|
Sakai A, Yasui T, Watanave M, Tatsumi R, Yamamoto Y, Takano W, Tani Y, Okamura I, Hirai H, Takeda S. Development of novel potent ligands for GPR85, an orphan G protein-coupled receptor expressed in the brain. Genes Cells 2022; 27:345-355. [PMID: 35194900 DOI: 10.1111/gtc.12931] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 11/28/2022]
Abstract
GPR85 is a member of the G protein-coupled receptor and is a super-conserved receptor expressed in the brain sub-family (Super Conserved Receptor Expressed in Brain; SREB) with GPR27 and GPR173. These three receptors are "orphan receptors"; however, their endogenous ligands have not been identified. SREB has garnered the interest of many scientists because it is expressed in the central nervous system and is evolutionarily conserved. In particular, brain mass is reported to be increased and learning and memory are improved in GPR85 knockout mice (Matsumoto et al. 2008). In this study, we characterized newly synthesized compounds using a GPR85-Gsα fusion protein and the [35 S]GTPγS binding assay and identified novel GPR85 inverse-agonists with IC50 values of approximately 1 μM. To analyze the neurochemical character of the compounds and investigate the physiological significance of GPR85, we used cerebellar Purkinje cells expressing GPR85 and an electrophysiological technique. Based on the results, the inverse-agonist compound for GPR85 modulated potassium channel opening. Together with the results of previous gene analysis of GPR85, we expect that the development of the GPR85 ligand will provide new insights into a few types of neurological disorders.
Collapse
Affiliation(s)
- Aya Sakai
- Graduate of School of Science and Technology, Division of Molecular Science, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma, Japan
| | - Takeshi Yasui
- Graduate of School of Pharmaceutical Sciences, Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Chikusa, Nagoya, Japan
| | - Masashi Watanave
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, Japan
| | - Rine Tatsumi
- Graduate of School of Pharmaceutical Sciences, Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Chikusa, Nagoya, Japan
| | - Yoshihiko Yamamoto
- Graduate of School of Pharmaceutical Sciences, Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Chikusa, Nagoya, Japan
| | - Wataru Takano
- Graduate of School of Science and Technology, Division of Molecular Science, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma, Japan
| | - Yuki Tani
- Graduate of School of Science and Technology, Division of Molecular Science, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma, Japan
| | - Izumi Okamura
- Graduate of School of Science and Technology, Division of Molecular Science, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, Japan
| | - Shigeki Takeda
- Graduate of School of Science and Technology, Division of Molecular Science, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma, Japan
| |
Collapse
|
7
|
Mpoulimari I, Zintzaras E. Identification of Chromosomal Regions Linked to Autism-Spectrum Disorders: A Meta-Analysis of Genome-Wide Linkage Scans. Genet Test Mol Biomarkers 2022; 26:59-69. [PMID: 35225680 DOI: 10.1089/gtmb.2021.0236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background: Autism spectrum disorder (ASD) is a clinically and genetically heterogeneous group of pervasive neurodevelopmental disorders with a strong hereditary component. Although, genome-wide linkage scans (GWLS) and association studies (GWAS) have previously identified hundreds of ASD risk gene loci, the results remain inconclusive. Method: We performed a heterogeneity-based genome search meta-analysis (HEGESMA) of 15 genome scans of autism and ASD. Results: For strictly defined autism, data were analyzed across six separate genome scans. Region 7q22-q34 reached statistical significance in both weighted and unweighted analyses, with evidence of significantly low between-scan heterogeneity. For ASDs (data from 12 separate scans), chromosomal regions 5p15.33-5p15.1 and 15q22.32-15q26.1 reached significance in both weighted and unweighted analyses but did not reach significance for either low or high heterogeneity. Region 1q23.2-1q31.1 was significant in unweighted analyses with low between-scan heterogeneity. Finally, region 8p21.1-8q13.2 reached significant linkage peak in all our meta-analyses. When we combined all available genome scans (15), the same results were produced. Conclusions: This meta-analysis suggests that these regions should be further investigated for autism susceptibility genes, with the caveat that autism spectrum disorders have different linkage signals across genome scans, possibly because of the high genetic heterogeneity of the disease.
Collapse
Affiliation(s)
- Ioanna Mpoulimari
- Department of Biomathematics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Elias Zintzaras
- Department of Biomathematics, Faculty of Medicine, University of Thessaly, Larissa, Greece
- The Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Rikos D, Siokas V, Burykina TI, Drakoulis N, Dardiotis E, Zintzaras E. Replication of chromosomal loci involved in Parkinson's disease: A quantitative synthesis of GWAS. Toxicol Rep 2021; 8:1762-1768. [PMID: 34712594 PMCID: PMC8528647 DOI: 10.1016/j.toxrep.2021.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/15/2021] [Accepted: 10/09/2021] [Indexed: 11/16/2022] Open
Abstract
The first quantitative synthesis of GWAS regarding Parkinson’s Disease. Fifteen Parkinson’s Disease GWASs with 191.397 available SNPs pooled. User friendly software (METRADISC-XL) implemented. Seven chromosomal regions (bins) were replicated as associated with the Parkinson’s Disease trait.
Introduction Parkinson’s disease is a neurodegenerative disorder with a complex etiology coming from interactions between genetic and environmental factors. Research on Parkinson’s disease genetics has been an effortful struggle, while new technologies and novel study designs served as indispensable boosters. Until now, 90 loci and 20 disease-causing gene mutations have been identified. In this study we describe a novel non-parametric approach to GWAS meta-analysis and its application in PD genetics. Methods A literature search was conducted to identify Genome-Wide Association Studies (GWAS) regarding Parkinson’s disease. We applied predefined inclusion criteria and extracted the reported SNPs and their respective position and statistical significance. We divided all chromosomes in approximately equal genetic distance segments called bins and recorded the most significant SNP from each bin and each study and ranked them in terms of their p-value. Ranks from each bin were summed, averaged and added in a heterogeneity-based analysis using the METRADISC-XL software. Weighted and unweighted analysis was performed. Results Five-hundred and forty-three SNPs and their respective p-values from 15 studies were matched in their corresponding bins. The METRADISC-XL analysis resulted in 7 bins with a significant p-value. A bin on chromosome 4 where the SNCA gene is located found with genome-wide significant association with Parkinson’s Disease. Conclusion This is the first time a non-parametric method is applied in GWAS meta-analysis. The results add some insight on the overall understanding of Parkinson’s disease genetics and serve as a first step of further convergent analysis with Genome-wide linkage studies.
Collapse
Affiliation(s)
- Dimitrios Rikos
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, Larissa, Greece.,Department of Biomathematics, Faculty of Medicine, University of Thessaly Larissa, Greece
| | - Vasileios Siokas
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Tatyana I Burykina
- Department of Analytical and Forensic Medical Toxicology, Sechenov University, 119048 Moscow, Russian Federation
| | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 17551 Athens, Greece
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Elias Zintzaras
- Department of Biomathematics, Faculty of Medicine, University of Thessaly Larissa, Greece.,The Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
9
|
Uddin MG, Siddiqui SA, Uddin MS, Aziz MA, Hussain MS, Furhatun-Noor, Millat MS, Sen N, Muhuri B, Islam MS. Genetic variants of ZNF385B and COMT are associated with autism spectrum disorder in the Bangladeshi children. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
10
|
Andrade A, Brennecke A, Mallat S, Brown J, Gomez-Rivadeneira J, Czepiel N, Londrigan L. Genetic Associations between Voltage-Gated Calcium Channels and Psychiatric Disorders. Int J Mol Sci 2019; 20:E3537. [PMID: 31331039 PMCID: PMC6679227 DOI: 10.3390/ijms20143537] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/23/2022] Open
Abstract
Psychiatric disorders are mental, behavioral or emotional disorders. These conditions are prevalent, one in four adults suffer from any type of psychiatric disorders world-wide. It has always been observed that psychiatric disorders have a genetic component, however, new methods to sequence full genomes of large cohorts have identified with high precision genetic risk loci for these conditions. Psychiatric disorders include, but are not limited to, bipolar disorder, schizophrenia, autism spectrum disorder, anxiety disorders, major depressive disorder, and attention-deficit and hyperactivity disorder. Several risk loci for psychiatric disorders fall within genes that encode for voltage-gated calcium channels (CaVs). Calcium entering through CaVs is crucial for multiple neuronal processes. In this review, we will summarize recent findings that link CaVs and their auxiliary subunits to psychiatric disorders. First, we will provide a general overview of CaVs structure, classification, function, expression and pharmacology. Next, we will summarize tools to study risk loci associated with psychiatric disorders. We will examine functional studies of risk variations in CaV genes when available. Finally, we will review pharmacological evidence of the use of CaV modulators to treat psychiatric disorders. Our review will be of interest for those studying pathophysiological aspects of CaVs.
Collapse
Affiliation(s)
- Arturo Andrade
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA.
| | - Ashton Brennecke
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Shayna Mallat
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Julian Brown
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | | | - Natalie Czepiel
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Laura Londrigan
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| |
Collapse
|
11
|
Tziastoudi M, Stefanidis I, Stravodimos K, Zintzaras E. Identification of Chromosomal Regions Linked to Diabetic Nephropathy: A Meta-Analysis of Genome-Wide Linkage Scans. Genet Test Mol Biomarkers 2019; 23:105-117. [DOI: 10.1089/gtmb.2018.0209] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Maria Tziastoudi
- Department of Biomathematics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Konstantinos Stravodimos
- 1st University Department of Urology, Laiko General Hospital, National and Kapodistrian Athens University, Athens, Greece
| | - Elias Zintzaras
- Department of Biomathematics, Faculty of Medicine, University of Thessaly, Larissa, Greece
- The Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | | |
Collapse
|
12
|
Opposite development of short- and long-range anterior cingulate pathways in autism. Acta Neuropathol 2018; 136:759-778. [PMID: 30191402 PMCID: PMC6208731 DOI: 10.1007/s00401-018-1904-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/27/2018] [Accepted: 08/29/2018] [Indexed: 12/31/2022]
Abstract
Autism has been linked with the changes in brain connectivity that disrupt neural communication, especially involving frontal networks. Pathological changes in white matter are evident in adults with autism, particularly affecting axons below the anterior cingulate cortices (ACC). It is still unknown whether axon pathology appears early or late in development and whether it changes or not from childhood through adulthood. To address these questions, we examined typical and pathological development of about 1 million axons in post-mortem brains of children, adolescents, and adults with and without autism (ages 3-67 years). We used high-resolution microscopy to systematically sample and study quantitatively the fine structure of myelinated axons in the white matter below ACC. We provide novel evidence of changes in the density, size and trajectories of ACC axons in typical postnatal development from childhood through adulthood. Against the normal profile of axon development, our data revealed lower density of myelinated axons that connect ACC with neighboring cortices in children with autism. In the course of development the proportion of thin axons, which form short-range pathways, increased significantly in individuals with autism, but remained flat in controls. In contrast, the relative proportion of thick axons, which form long-range pathways, increased from childhood to adulthood in the control group, but decreased in autism. Our findings provide a timeline for profound changes in axon density and thickness below ACC that affect axon physiology in a direction suggesting bias in short over distant neural communication in autism. Importantly, measures of axon density, myelination, and orientation provide white matter anisotropy/diffusivity estimates at the level of single axons. The structural template established can be used to compare with measures obtained from imaging in living subjects, and guide analysis of functional and structural imaging data from humans for comparison with pathological states.
Collapse
|
13
|
Gross C. Defective phosphoinositide metabolism in autism. J Neurosci Res 2016; 95:1161-1173. [PMID: 27376697 DOI: 10.1002/jnr.23797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/26/2016] [Accepted: 06/01/2016] [Indexed: 12/12/2022]
Abstract
Phosphoinositides are essential components of lipid membranes and crucial regulators of many cellular functions, including signal transduction, vesicle trafficking, membrane receptor localization and activity, and determination of membrane identity. These functions depend on the dynamic and highly regulated metabolism of phosphoinositides and require finely balanced activity of specific phosphoinositide kinases and phosphatases. There is increasing evidence from genetic and functional studies that these enzymes are often dysregulated or mutated in autism spectrum disorders; in particular, phosphoinositide 3-kinases and their regulatory subunits appear to be affected frequently. Examples of autism spectrum disorders with defective phosphoinositide metabolism are fragile X syndrome and autism disorders associated with mutations in the phosphoinositide 3-phosphatase tensin homolog deleted on chromosome 10 (PTEN), but recent genetic analyses also suggest that select nonsyndromic, idiopathic forms of autism may have altered activity of phosphoinositide kinases and phosphatases. Isoform-specific inhibitors for some of the phosphoinositide kinases have already been developed for cancer research and treatment, and a few are being evaluated for use in humans. Altogether, this offers exciting opportunities to explore altered phosphoinositide metabolism as a therapeutic target in individuals with certain forms of autism. This review summarizes genetic and functional studies identifying defects in phosphoinositide metabolism in autism and related disorders, describes published preclinical work targeting phosphoinositide 3-kinases in neurological diseases, and discusses the opportunities and challenges ahead to translate these findings from animal models and human cells into clinical application in humans. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
14
|
Merikangas AK, Segurado R, Heron EA, Anney RJL, Paterson AD, Cook EH, Pinto D, Scherer SW, Szatmari P, Gill M, Corvin AP, Gallagher L. The phenotypic manifestations of rare genic CNVs in autism spectrum disorder. Mol Psychiatry 2015; 20:1366-72. [PMID: 25421404 PMCID: PMC4759095 DOI: 10.1038/mp.2014.150] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 08/10/2014] [Accepted: 09/19/2014] [Indexed: 12/28/2022]
Abstract
Significant evidence exists for the association between copy number variants (CNVs) and Autism Spectrum Disorder (ASD); however, most of this work has focused solely on the diagnosis of ASD. There is limited understanding of the impact of CNVs on the 'sub-phenotypes' of ASD. The objective of this paper is to evaluate associations between CNVs in differentially brain expressed (DBE) genes or genes previously implicated in ASD/intellectual disability (ASD/ID) and specific sub-phenotypes of ASD. The sample consisted of 1590 cases of European ancestry from the Autism Genome Project (AGP) with a diagnosis of an ASD and at least one rare CNV impacting any gene and a core set of phenotypic measures, including symptom severity, language impairments, seizures, gait disturbances, intelligence quotient (IQ) and adaptive function, as well as paternal and maternal age. Classification analyses using a non-parametric recursive partitioning method (random forests) were employed to define sets of phenotypic characteristics that best classify the CNV-defined groups. There was substantial variation in the classification accuracy of the two sets of genes. The best variables for classification were verbal IQ for the ASD/ID genes, paternal age at birth for the DBE genes and adaptive function for de novo CNVs. CNVs in the ASD/ID list were primarily associated with communication and language domains, whereas CNVs in DBE genes were related to broader manifestations of adaptive function. To our knowledge, this is the first study to examine the associations between sub-phenotypes and CNVs genome-wide in ASD. This work highlights the importance of examining the diverse sub-phenotypic manifestations of CNVs in ASD, including the specific features, comorbid conditions and clinical correlates of ASD that comprise underlying characteristics of the disorder.
Collapse
Affiliation(s)
- A K Merikangas
- Department of Psychiatry, Neuropsychiatric Genetics Research Group, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland,Department of Psychiatry, Neuropsychiatric Genetics Research Group, Trinity College Dublin, Institute of Molecular Medicine, St. James's Hospital, James's Street, Dublin, Dublin 8, Ireland. E-mail:
| | - R Segurado
- Centre for Support and Training in Analysis and Research, University College Dublin, Dublin 4, Ireland
| | - E A Heron
- Department of Psychiatry, Neuropsychiatric Genetics Research Group, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - R J L Anney
- Department of Psychiatry, Neuropsychiatric Genetics Research Group, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - A D Paterson
- Program in Genetics and Genome Biology, Hospital for Sick Children, Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - E H Cook
- Institute for Juvenile Research, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - D Pinto
- Departments of Psychiatry, and Genetics and Genomic Sciences, Seaver Autism Center, The Mindich Child Health & Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - S W Scherer
- Department of Molecular Genetics, The Centre for Applied Genomics and Program in Genetics and Genomic Biology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - P Szatmari
- The Division of Child and Adolescent Psychiatry, Centre for Addiction and Mental Health, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - M Gill
- Department of Psychiatry, Neuropsychiatric Genetics Research Group, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - A P Corvin
- Department of Psychiatry, Neuropsychiatric Genetics Research Group, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - L Gallagher
- Department of Psychiatry, Neuropsychiatric Genetics Research Group, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| |
Collapse
|
15
|
Liang S, Wang XL, Zou MY, Wang H, Zhou X, Sun CH, Xia W, Wu LJ, Fujisawa TX, Tomoda A. Family-based association study of ZNF533, DOCK4 and IMMP2L gene polymorphisms linked to autism in a northeastern Chinese Han population. J Zhejiang Univ Sci B 2014; 15:264-71. [PMID: 24599690 DOI: 10.1631/jzus.b1300133] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE A study in a Caucasian population has identified two single-nucleotide polymorphisms (SNPs) in ZNF533, one in DOCK4, and two in IMMP2L, which were all significantly associated with autism. They are located in AUTS1 and AUTS5, which have been identified as autism susceptibility loci in several genome-wide screens. The present study aimed to investigate whether ZNF533, DOCK4, and IMMP2L genes are also associated with autism in a northeastern Chinese Han population. METHODS We performed a similar association study using families with three individuals (one autistic child and two unaffected parents). A family-based transmission disequilibrium test (TDT) was used to analyze the results. RESULTS There were significant associations between autism and the two SNPs of ZNF533 gene (rs11885327: χ(2)=4.5200, P=0.0335; rs1964081: χ(2)=4.2610, P=0.0390) and the SNP of DOCK4 gene (rs2217262: χ(2)=5.3430, P=0.0208). CONCLUSIONS Our data suggest that ZNF533 and DOCK4 genes are linked to a predisposition to autism in the northeastern Chinese Han population.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China; Research Center for Child Mental Development, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji-cho, Fukui 910-1193, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Sánchez Delgado M, Camprubí C, Tümer Z, Martínez F, Milà M, Monk D. Screening individuals with intellectual disability, autism and Tourette's syndrome for KCNK9 mutations and aberrant DNA methylation within the 8q24 imprinted cluster. Am J Med Genet B Neuropsychiatr Genet 2014; 165B:472-8. [PMID: 24980697 DOI: 10.1002/ajmg.b.32250] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/27/2014] [Indexed: 12/22/2022]
Abstract
The phenotype overlap between autism spectrum disorders (ASD) & intellectual disabilities (ID) is mirrored at the genetic level, with common genes being reported mutated in variety of developmental disabilities. However despite widespread genetic screening for mutations, in approximately 40-60% of childhood developmental disorders the genetic cause remains unknown. Several genome-wide linkage screens in ASD have identified a locus mapping to distal 8q. We have recently identified a novel brain-specific imprinted cluster at this location, which contains the reciprocally expressed maternal KCNK9 and paternally expressed non-coding PEG13 transcripts, the latter located within an intron of TRAPPC9. Interestingly, mutations of KCNK9 and TRAPPC9 have been reported in Birk-Barel mental retardation and non-syndromic familial forms of ID, respectively. Here, we report a genetic screen for KCNK9 coding mutations and potential epigenetic aberrations that could result in deregulated imprinting in a cohort of 120 ID, 86 ASD and 86 Tourette syndrome patients. Fifteen of the ID patients had clinical characteristics overlapping with Birk-Barel syndrome. Sequencing of the two coding exons of KCNK9 failed to identify pathologic mutations, with only one variant, rs2615374, being present with allele frequencies similar to those described in dbSNP database. DNA methylation profiling of the KCNK9 and TRAPPC9 promoters, the maternally methylated PEG13 DMR and a long-range enhancer region were normal in all patients. Our findings suggest that mutations of KCNK9 or epigenetic disturbances within the PEG13 imprinted cluster do not significantly contribute to the cause of the developmental disabilities tested in this study.
Collapse
Affiliation(s)
- Marta Sánchez Delgado
- Imprinting and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
17
|
Breitenkamp AFS, Matthes J, Nass RD, Sinzig J, Lehmkuhl G, Nürnberg P, Herzig S. Rare mutations of CACNB2 found in autism spectrum disease-affected families alter calcium channel function. PLoS One 2014; 9:e95579. [PMID: 24752249 PMCID: PMC3994086 DOI: 10.1371/journal.pone.0095579] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 03/27/2014] [Indexed: 01/06/2023] Open
Abstract
Autism Spectrum Disorders (ASD) are complex neurodevelopmental diseases clinically defined by dysfunction of social interaction. Dysregulation of cellular calcium homeostasis might be involved in ASD pathogenesis, and genes coding for the L-type calcium channel subunits CaV1.2 (CACNA1C) and CaVβ2 (CACNB2) were recently identified as risk loci for psychiatric diseases. Here, we present three rare missense mutations of CACNB2 (G167S, S197F, and F240L) found in ASD-affected families, two of them described here for the first time (G167S and F240L). All these mutations affect highly conserved regions while being absent in a sample of ethnically matched controls. We suggest the mutations to be of physiological relevance since they modulate whole-cell Ba2+ currents through calcium channels when expressed in a recombinant system (HEK-293 cells). Two mutations displayed significantly decelerated time-dependent inactivation as well as increased sensitivity of voltage-dependent inactivation. In contrast, the third mutation (F240L) showed significantly accelerated time-dependent inactivation. By altering the kinetic parameters, the mutations are reminiscent of the CACNA1C mutation causing Timothy Syndrome, a Mendelian disease presenting with ASD. In conclusion, the results of our first-time biophysical characterization of these three rare CACNB2 missense mutations identified in ASD patients support the hypothesis that calcium channel dysfunction may contribute to autism.
Collapse
Affiliation(s)
| | - Jan Matthes
- Department of Pharmacology, University of Cologne, Cologne, Germany
| | | | - Judith Sinzig
- Department of Child and Adolescent Psychiatry and Psychotherapy, LVR-Klinik Bonn, Bonn, Germany
- Department of Child and Adolescent Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Gerd Lehmkuhl
- Department of Child and Adolescent Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Stefan Herzig
- Department of Pharmacology, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
- * E-mail:
| |
Collapse
|
18
|
Herbert MR, Sage C. Autism and EMF? Plausibility of a pathophysiological link - Part I. ACTA ACUST UNITED AC 2013; 20:191-209. [PMID: 24095003 DOI: 10.1016/j.pathophys.2013.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 05/06/2013] [Accepted: 07/15/2013] [Indexed: 01/02/2023]
Abstract
Although autism spectrum conditions (ASCs) are defined behaviorally, they also involve multileveled disturbances of underlying biology that find striking parallels in the physiological impacts of electromagnetic frequency and radiofrequency exposures (EMF/RFR). Part I of this paper will review the critical contributions pathophysiology may make to the etiology, pathogenesis and ongoing generation of core features of ASCs. We will review pathophysiological damage to core cellular processes that are associated both with ASCs and with biological effects of EMF/RFR exposures that contribute to chronically disrupted homeostasis. Many studies of people with ASCs have identified oxidative stress and evidence of free radical damage, cellular stress proteins, and deficiencies of antioxidants such as glutathione. Elevated intracellular calcium in ASCs may be due to genetics or may be downstream of inflammation or environmental exposures. Cell membrane lipids may be peroxidized, mitochondria may be dysfunctional, and various kinds of immune system disturbances are common. Brain oxidative stress and inflammation as well as measures consistent with blood-brain barrier and brain perfusion compromise have been documented. Part II of this paper will review how behaviors in ASCs may emerge from alterations of electrophysiological oscillatory synchronization, how EMF/RFR could contribute to these by de-tuning the organism, and policy implications of these vulnerabilities. Changes in brain and autonomic nervous system electrophysiological function and sensory processing predominate, seizures are common, and sleep disruption is close to universal. All of these phenomena also occur with EMF/RFR exposure that can add to system overload ('allostatic load') in ASCs by increasing risk, and worsening challenging biological problems and symptoms; conversely, reducing exposure might ameliorate symptoms of ASCs by reducing obstruction of physiological repair. Various vital but vulnerable mechanisms such as calcium channels may be disrupted by environmental agents, various genes associated with autism or the interaction of both. With dramatic increases in reported ASCs that are coincident in time with the deployment of wireless technologies, we need aggressive investigation of potential ASC - EMF/RFR links. The evidence is sufficient to warrant new public exposure standards benchmarked to low-intensity (non-thermal) exposure levels now known to be biologically disruptive, and strong, interim precautionary practices are advocated.
Collapse
Affiliation(s)
- Martha R Herbert
- TRANSCEND Research Program Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA.
| | | |
Collapse
|
19
|
Abstract
Developing new pharmacotherapies for autism spectrum disorder (ASD) is a challenge. ASD has a complex genetic architecture, several neurobiological phenotypes and multiple symptom domains. However, new opportunities are emerging that could lead to the development of 'targeted' and individualized pharmacological interventions. Here, first we review these important new insights into the aetiology and neurobiology of ASD with particular focus on (i) genetic variants mediating synaptic structure and functioning and (ii) differences in brain anatomy, chemistry and connectivity in this condition. The characterization of the genotypic and phenotypic differences underlying ASD might in the future be invaluable for stratifying the large range of different individuals on the autism spectrum into genetically and/or biologically homogeneous subgroups that might respond to similar targeted interventions. Secondly, we propose a strategic framework for the development of targeted pharmacotherapies for ASD, which comprises several different stages in which research findings are translated into clinical applications. The establishment of animal models and cellular assays is important for developing and testing new pharmacological targets before initiating large-scale clinical trials. Finally, we present the European Autism Interventions - A Multicentre Study for Developing New Medications (EU-AIMS) Initiative, which was set up in the context of the EU Innovative Medicines Initiative as the first European platform for integrated translational research in ASD. The EU-AIMS Initiative consists of academic and industrial partners working in collaboration to deliver a more 'personalized' approach to diagnosing and treating ASD in the future.
Collapse
Affiliation(s)
- C Ecker
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, London, UK
| | | | | |
Collapse
|
20
|
Zikopoulos B, Barbas H. Altered neural connectivity in excitatory and inhibitory cortical circuits in autism. Front Hum Neurosci 2013; 7:609. [PMID: 24098278 PMCID: PMC3784686 DOI: 10.3389/fnhum.2013.00609] [Citation(s) in RCA: 209] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 09/06/2013] [Indexed: 12/12/2022] Open
Abstract
Converging evidence from diverse studies suggests that atypical brain connectivity in autism affects in distinct ways short- and long-range cortical pathways, disrupting neural communication and the balance of excitation and inhibition. This hypothesis is based mostly on functional non-invasive studies that show atypical synchronization and connectivity patterns between cortical areas in children and adults with autism. Indirect methods to study the course and integrity of major brain pathways at low resolution show changes in fractional anisotropy (FA) or diffusivity of the white matter in autism. Findings in post-mortem brains of adults with autism provide evidence of changes in the fine structure of axons below prefrontal cortices, which communicate over short- or long-range pathways with other cortices and subcortical structures. Here we focus on evidence of cellular and axon features that likely underlie the changes in short- and long-range communication in autism. We review recent findings of changes in the shape, thickness, and volume of brain areas, cytoarchitecture, neuronal morphology, cellular elements, and structural and neurochemical features of individual axons in the white matter, where pathology is evident even in gross images. We relate cellular and molecular features to imaging and genetic studies that highlight a variety of polymorphisms and epigenetic factors that primarily affect neurite growth and synapse formation and function in autism. We report preliminary findings of changes in autism in the ratio of distinct types of inhibitory neurons in prefrontal cortex, known to shape network dynamics and the balance of excitation and inhibition. Finally we present a model that synthesizes diverse findings by relating them to developmental events, with a goal to identify common processes that perturb development in autism and affect neural communication, reflected in altered patterns of attention, social interactions, and language.
Collapse
Affiliation(s)
- Basilis Zikopoulos
- Neural Systems Laboratory, Department of Health Sciences, Boston University Boston, MA, USA
| | | |
Collapse
|
21
|
Ecker C, Spooren W, Murphy DGM. Translational approaches to the biology of Autism: false dawn or a new era? Mol Psychiatry 2013; 18:435-42. [PMID: 22801412 PMCID: PMC3606942 DOI: 10.1038/mp.2012.102] [Citation(s) in RCA: 68] [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: 02/22/2012] [Revised: 05/29/2012] [Accepted: 05/31/2012] [Indexed: 02/04/2023]
Abstract
Discovering novel treatments for Autism Spectrum Disorders (ASD) is a challenge. Its etiology and pathology remain largely unknown, the condition shows wide clinical diversity, and case identification is still solely based on symptomatology. Hence clinical trials typically include samples of biologically and clinically heterogeneous individuals. 'Core deficits', that is, deficits common to all individuals with ASD, are thus inherently difficult to find. Nevertheless, recent reports suggest that new opportunities are emerging, which may help develop new treatments and biomarkers for the condition. Most important, several risk gene variants have now been identified that significantly contribute to ASD susceptibility, many linked to synaptic functioning, excitation-inhibition balance, and brain connectivity. Second, neuroimaging studies have advanced our understanding of the 'wider' neural systems underlying ASD; and significantly contributed to our knowledge of the complex neurobiology associated with the condition. Last, the recent development of powerful multivariate analytical techniques now enable us to use multi-modal information in order to develop complex 'biomarker systems', which may in the future be used to assist the behavioral diagnosis, aid patient stratification and predict response to treatment/intervention. The aim of this review is, therefore, to summarize some of these important new findings and highlight their potential significant translational value to the future of ASD research.
Collapse
Affiliation(s)
- C Ecker
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, King's College, London, UK.
| | | | | |
Collapse
|
22
|
Radulescu E, Sambataro F, Mattay VS, Callicott JH, Straub RE, Matsumoto M, Weinberger DR, Marenco S. Effect of schizophrenia risk-associated alleles in SREB2 (GPR85) on functional MRI phenotypes in healthy volunteers. Neuropsychopharmacology 2013; 38:341-9. [PMID: 22968816 PMCID: PMC3527120 DOI: 10.1038/npp.2012.184] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 08/06/2012] [Accepted: 08/07/2012] [Indexed: 01/08/2023]
Abstract
Genetic variants in GPR85 (SREB2: rs56080411 and rs56039557) have been associated with risk for schizophrenia. Here, we test the hypothesis that these variants impact on brain function in normal subjects, measured with functional magnetic resonance imaging (fMRI) paradigms that target regions with greatest SREB2 expression (hippocampal formation and amygdaloid complex). During a facial emotion recognition paradigm, a significant interaction of rs56080411 genotype by sex was found in the left amygdaloid complex (male risk allele carriers showed less activation than male homozygotes for the non-risk allele, while females showed the opposite pattern). During aversive encoding of an emotional memory paradigm, we found that risk allele carriers for rs56080411 had greater activation in the right inferior frontal gyrus. Trends in the same direction were present for rs56039557 in the right occipital cortex and right fusiform gyrus. During a working memory paradigm, a significant sex-by-genotype interaction was found with male risk allele carriers of rs56080411 having inefficient activation within the left dorsolateral prefrontal cortex (DLPFC), compared with same sex non-risk carriers, while females revealed an opposite pattern, despite similar levels of performance. These data suggest that risk-associated variants in SREB2 are associated with phenotypes similar to those found in patients with schizophrenia in the DLPFC and the amygdala of males, while the pattern is opposite in females. The findings in females and during the emotional memory paradigm are consistent with modulation by SREB2 of brain circuitries implicated in mood regulation and may be relevant to neuropsychiatric conditions other than schizophrenia.
Collapse
Affiliation(s)
| | - Fabio Sambataro
- Clinical Brain Disorders Branch, GCAP, IRP,
NIMH, Bethesda, MD, USA
| | - Venkata S Mattay
- Clinical Brain Disorders Branch, GCAP, IRP,
NIMH, Bethesda, MD, USA
| | | | - Richard E Straub
- Clinical Brain Disorders Branch, GCAP, IRP,
NIMH, Bethesda, MD, USA
| | | | | | - Stefano Marenco
- Clinical Brain Disorders Branch, GCAP, IRP,
NIMH, Bethesda, MD, USA
| |
Collapse
|
23
|
Abstract
Diabetic nephropathy (DN) is a devastating complication of type 1 and type 2 diabetes and leads to increased morbidity and premature mortality. Susceptibility to DN has an inherent genetic basis as evidenced by familial aggregation and ethnic-specific prevalence rates. Progress in identifying the underlying genetic architecture has been arduous with the realization that a single locus of large effect does not exist, unlike in predisposition to non-diabetic nephropathy in individuals with African ancestry. Numerous risk variants have been identified, each with a nominal effect, and they collectively contribute to disease. These results have identified loci targeting novel pathways for disease susceptibility. With continued technological advances and development of new analytic methods, additional genetic variants and mechanisms (e.g., epigenetic variation) will be identified and help to elucidate the pathogenesis of DN. These advances will lead to early detection and development of novel therapeutic strategies to decrease the incidence of disease.
Collapse
Affiliation(s)
- Nicholette D. Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston Salem, NC USA
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston Salem, NC USA
| | - Barry I. Freedman
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston Salem, NC USA
| |
Collapse
|
24
|
Sullivan PF, Daly MJ, O'Donovan M. Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat Rev Genet 2012; 13:537-51. [PMID: 22777127 PMCID: PMC4110909 DOI: 10.1038/nrg3240] [Citation(s) in RCA: 842] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Psychiatric disorders are among the most intractable enigmas in medicine. In the past 5 years, there has been unprecedented progress on the genetics of many of these conditions. In this Review, we discuss the genetics of nine cardinal psychiatric disorders (namely, Alzheimer's disease, attention-deficit hyperactivity disorder, alcohol dependence, anorexia nervosa, autism spectrum disorder, bipolar disorder, major depressive disorder, nicotine dependence and schizophrenia). Empirical approaches have yielded new hypotheses about aetiology and now provide data on the often debated genetic architectures of these conditions, which have implications for future research strategies. Further study using a balanced portfolio of methods to assess multiple forms of genetic variation is likely to yield many additional new findings.
Collapse
Affiliation(s)
- Patrick F Sullivan
- Departments of Genetics and Psychiatry, CB# 7264, 5097 Genomic Medicine, University of North Carolina at Chapel Hill, North Carolina 27599-27264, USA.
| | | | | |
Collapse
|
25
|
Freitag CM, Asherson P, Hebebrand J. Behavioural genetics of childhood disorders. Curr Top Behav Neurosci 2012; 12:395-428. [PMID: 22382729 DOI: 10.1007/7854_2011_178] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
After a general introduction into genetic risk factors for child psychiatric disorders, four specific child psychiatric disorders with a strong genetic component, namely, Autism Spectrum Disorders, Attention Deficit / Hyperactivity Disorder, Nocturnal Enuresis, and obesity, are discussed in detail. Recent evidence of linkage, candidate gene, and genome-wide association studies are presented. This chapter ends with a prospectus on further research needs.
Collapse
Affiliation(s)
- Christine M Freitag
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe-Universität Frankfurt am Main, Deutschordenstraße 50, 60528, Frankfurt am Main, Germany,
| | | | | |
Collapse
|
26
|
De Rubeis S, Bagni C. Regulation of molecular pathways in the Fragile X Syndrome: insights into Autism Spectrum Disorders. J Neurodev Disord 2011; 3:257-69. [PMID: 21842222 PMCID: PMC3167042 DOI: 10.1007/s11689-011-9087-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Accepted: 07/07/2011] [Indexed: 11/01/2022] Open
Abstract
The Fragile X syndrome (FXS) is a leading cause of intellectual disability (ID) and autism. The disease is caused by mutations or loss of the Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein playing multiple functions in RNA metabolism. The expression of a large set of neuronal mRNAs is altered when FMRP is lost, thus causing defects in neuronal morphology and physiology. FMRP regulates mRNA stability, dendritic targeting, and protein synthesis. At synapses, FMRP represses protein synthesis by forming a complex with the Cytoplasmic FMRP Interacting Protein 1 (CYFIP1) and the cap-binding protein eIF4E. Here, we review the clinical, genetic, and molecular aspects of FXS with a special focus on the receptor signaling that regulates FMRP-dependent protein synthesis. We further discuss the FMRP-CYFIP1 complex and its potential relevance for ID and autism.
Collapse
Affiliation(s)
- Silvia De Rubeis
- Center for Human Genetics, Katholieke Universiteit Leuven, 3000, Leuven, Belgium
| | | |
Collapse
|
27
|
Association study of the CNS patterning genes and autism in Han Chinese in Taiwan. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:1512-7. [PMID: 21575668 DOI: 10.1016/j.pnpbp.2011.04.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 04/18/2011] [Accepted: 04/25/2011] [Indexed: 10/18/2022]
Abstract
Autism is a complex neurodevelopmental disorder with high heritability. Despite different approaches worldwide to identify susceptibility loci or genes for autism spectrum disorders (ASDs), no consistent result has been reported. CNS patterning genes have been recognized as candidate genes for autism based on neuroimage and neuropathology evidence. This study investigated four candidate genes (WNT2, EN2, SHANK3, and FOXP2) by a tag SNP approach in a family-based association study. The trio samples include 1164 subjects from 393 families, including 393 probands (aged 9.1±4.0years; male, 88.6%) diagnosed with autistic disorder (n=373) or Asperger's disorder (n=20) according to the DSM-IV diagnostic criteria and confirmed by the Chinese ADI-R interview. Three tag SNPs of EN2 (7q36), 6 SNPs of WNT2 (7q31-33), 5 SNPs of SHANK3 (22q13.3), 3 SNPs of FOXP2 (7q31) were genotyped. TDT analysis was done to test the association of each tag SNP and haplotype. There was no association with autism for 17 tag SNPs of WNT2, EN2, SHANK3, and FOXP2 based on SNP analyses. Haplotype analyses did not reveal significant association except for the 6 tag SNPs of WNT2 gene showing a significant association on one haplotype composed of rs2896218 and rs6950765 (G-G) (p=0.0095). Other haplotypes composed of rs2896218 and rs6950765 (G-G) were also significantly associated with autism. The present study indicates that SHANK3 may not be a critical gene for the etiology of ASDs in Han Chinese population. Inconsistent findings in EN2 and FOXP2 in the Han Chinese population need further clarification. A haplotype of WNT2 (rs2896218-rs6950765: G-G) is significantly associated with ASDs in our trios samples, this finding warrants further validation by different sample and confirmation by functional study.
Collapse
|
28
|
Liu XQ, Georgiades S, Duku E, Thompson A, Devlin B, Cook EH, Wijsman EM, Paterson AD, Szatmari P. Identification of genetic loci underlying the phenotypic constructs of autism spectrum disorders. J Am Acad Child Adolesc Psychiatry 2011; 50:687-696.e13. [PMID: 21703496 PMCID: PMC3593812 DOI: 10.1016/j.jaac.2011.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 05/05/2011] [Accepted: 05/09/2011] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To investigate the underlying phenotypic constructs in autism spectrum disorders (ASD) and to identify genetic loci that are linked to these empirically derived factors. METHOD Exploratory factor analysis was applied to two datasets with 28 selected Autism Diagnostic Interview-Revised (ADI-R) algorithm items. The first dataset was from the Autism Genome Project (AGP) phase I (1,236 ASD subjects from 618 families); the second was from the AGP phase II (804 unrelated ASD subjects). Variables derived from the factor analysis were then used as quantitative traits in genome-wide variance components linkage analyses. RESULTS Six factors, namely, joint attention, social interaction and communication, nonverbal communication, repetitive sensory-motor behavior, peer interaction, and compulsion/restricted interests, were retained for both datasets. There was good agreement between the factor loading patterns from the two datasets. All factors showed familial aggregation. Suggestive evidence for linkage was obtained for the joint attention factor on 11q23. Genome-wide significant evidence for linkage was obtained for the repetitive sensory-motor behavior factor on 19q13.3. CONCLUSIONS This study demonstrates that the underlying phenotypic constructs based on the ADI-R algorithm items are replicable in independent datasets, and that the empirically derived factors are suitable and informative in genetic studies of ASD.
Collapse
|
29
|
Rao M, Mottl AK, Cole SA, Umans JG, Freedman BI, Bowden DW, Langefeld CD, Fox CS, Yang Q, Cupples A, Iyengar SK, Hunt SC, Trikalinos TA. Meta-analysis of genome-wide linkage scans for renal function traits. Nephrol Dial Transplant 2011; 27:647-56. [PMID: 21622988 DOI: 10.1093/ndt/gfr255] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Several genome scans have explored the linkage of chronic kidney disease phenotypes to chromosomic regions with disparate results. Genome scan meta-analysis (GSMA) is a quantitative method to synthesize linkage results from independent studies and assess their concordance. METHODS We searched PubMed to identify genome linkage analyses of renal function traits in humans, such as estimated glomerular filtration rate (GFR), albuminuria, serum creatinine concentration and creatinine clearance. We contacted authors for numerical data and extracted information from individual studies. We applied the GSMA nonparametric approach to combine results across 14 linkage studies for GFR, 11 linkage studies for albumin creatinine ratio, 11 linkage studies for serum creatinine and 4 linkage studies for creatinine clearance. RESULTS No chromosomal region reached genome-wide statistical significance in the main analysis which included all scans under each phenotype; however, regions on Chromosomes 7, 10 and 16 reached suggestive significance for linkage to two or more phenotypes. Subgroup analyses by disease status or ethnicity did not yield additional information. CONCLUSIONS While heterogeneity across populations, methodologies and study designs likely explain this lack of agreement, it is possible that linkage scan methodologies lack the resolution for investigating complex traits. Combining family-based linkage studies with genome-wide association studies may be a powerful approach to detect private mutations contributing to complex renal phenotypes.
Collapse
Affiliation(s)
- Madhumathi Rao
- Division of Nephrology, Tufts Medical Center, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lack of Correlation Between Metallic Elements Analyzed in Hair by ICP-MS and Autism. J Autism Dev Disord 2011; 42:342-53. [DOI: 10.1007/s10803-011-1245-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
31
|
Korvatska O, Estes A, Munson J, Dawson G, Bekris LM, Kohen R, Yu CE, Schellenberg GD, Raskind WH. Mutations in the TSGA14 gene in families with autism spectrum disorders. Am J Med Genet B Neuropsychiatr Genet 2011; 156B:303-11. [PMID: 21438139 PMCID: PMC3552624 DOI: 10.1002/ajmg.b.31162] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 11/30/2010] [Indexed: 11/11/2022]
Abstract
Linkage to 7q has been the most robust genetic finding in familial autism. A previous scan of multiplex families with autism spectrum disorders found a linkage signal of genome-wide significance at D7S530 on 7q32. We searched a candidate imprinted region at this location for genetic variants in families with positive linkage scores. Using exon resequencing, we identified three rare potentially pathogenic variants in the TSGA14 gene, which encodes a centrosomal protein. Two variants were missense mutations (c.664C>G; p.P206A and c.766T>G; p.C240G) that changed conserved residues in the same protein domain; the third variant (c.192+5G>A) altered splicing, which resulted in a protein with an internal deletion of 16 residues and a G33D substitution. These rare TSGA14 variants are enriched in the affected subjects (6/348 patients versus 2/670 controls, Fisher's exact two tailed P = 0.022). This is the first report of a possible link of a gene with a centrosomal function with familial autism.
Collapse
Affiliation(s)
- O Korvatska
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, 98195, USA.
| | - A Estes
- Department of Speech and Hearing Sciences, University of Washington, Seattle, WA, USA
| | - J Munson
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - G Dawson
- Autism Speaks, New York, NY, USA,Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - L M Bekris
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - R Kohen
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - C-E Yu
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - G D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - W H Raskind
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
32
|
Guffanti G, Strik Lievers L, Bonati MT, Marchi M, Geronazzo L, Nardocci N, Estienne M, Larizza L, Macciardi F, Russo S. Role of UBE3A and ATP10A genes in autism susceptibility region 15q11-q13 in an Italian population: a positive replication for UBE3A. Psychiatry Res 2011; 185:33-8. [PMID: 20609483 DOI: 10.1016/j.psychres.2010.04.057] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 04/15/2010] [Accepted: 04/30/2010] [Indexed: 01/23/2023]
Abstract
The aetiology of autism is still largely unknown despite analyses from family and twin studies demonstrating substantial genetic role in the aetiology of the disorder. Data from linkage studies and analyses of chromosomal abnormalities identified 15q11-q13 as a region of particular aetiopathogenesis interest. We screened a set of markers spanning two known imprinted, maternally expressed genes, UBE3A and ATP10A, harboured in this candidate region. We replicated evidence of linkage disequilibrium (LD) at marker D15S122, located at the 5' end of UBE3A and originally reported by Nurmi et al. (2001). The potential role of UBE3A in our family-based association study is further supported by the association of two haplotypes that include one of the alleles of D15S122 and by the transmission disequilibrium test (TDT) evidence of the same allele in a parent of origin effect analysis. In a secondary analysis, we provided the first evidence of a significant association between first word delay and psychomotor regression with the 15q11-q13 region. Our data support a potential role of UBE3A in the complex pathogenic mechanisms of autism.
Collapse
|
33
|
Limited activity monitoring in toddlers with autism spectrum disorder. Brain Res 2010; 1380:246-54. [PMID: 21129365 DOI: 10.1016/j.brainres.2010.11.074] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 10/28/2010] [Accepted: 11/21/2010] [Indexed: 01/06/2023]
Abstract
This study used eye-tracking to examine how 20-month-old toddlers with autism spectrum disorder (ASD) (n=28), typical development (TD) (n=34), and non-autistic developmental delays (DD) (n=16) monitored the activities occurring in a context of an adult-child play interaction. Toddlers with ASD, in comparison to control groups, showed less attention to the activities of others and focused more on background objects (e.g., toys). In addition, while all groups spent the same time overall looking at people, toddlers with ASD looked less at people's heads and more at their bodies. In ASD, these patterns were associated with cognitive deficits and greater autism severity. These results suggest that the monitoring of the social activities of others is disrupted early in the developmental progression of autism, limiting future avenues for observational learning.
Collapse
|
34
|
Zaccaria KJ, Lagace DC, Eisch AJ, McCasland JS. Resistance to change and vulnerability to stress: autistic-like features of GAP43-deficient mice. GENES, BRAIN, AND BEHAVIOR 2010; 9:985-96. [PMID: 20707874 PMCID: PMC2975747 DOI: 10.1111/j.1601-183x.2010.00638.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
There is an urgent need for animal models of autism spectrum disorder (ASD) to understand the underlying pathology and facilitate development and testing of new treatments. The synaptic growth-associated protein-43 (GAP43) has recently been identified as an autism candidate gene of interest. Our previous studies show many brain abnormalities in mice lacking one allele for GAP43 [GAP43 (+/-)] that are consistent with the disordered connectivity theory of ASD. Thus, we hypothesized that GAP43 (+/-) mice would show at least some autistic-like behaviors. We found that GAP43 (+/-) mice, relative to wild-type (+/+) littermates, displayed resistance to change, consistent with one of the diagnostic criteria for ASD. GAP43 (+/-) mice also displayed stress-induced behavioral withdrawal and anxiety, as seen in many autistic individuals. In addition, both GAP43 (+/-) mice and (+/+) littermates showed low social approach and lack of preference for social novelty, consistent with another diagnostic criterion for ASD. This low sociability is likely because of the mixed C57BL/6J 129S3/SvImJ background. We conclude that GAP43 deficiency leads to the development of a subset of autistic-like behaviors. As these behaviors occur in a mouse that displays disordered connectivity, we propose that future anatomical and functional studies in this mouse may help uncover underlying mechanisms for these specific behaviors. Strain-specific low sociability may be advantageous in these studies, creating a more autistic-like environment for study of the GAP43-mediated deficits of resistance to change and vulnerability to stress.
Collapse
Affiliation(s)
- K J Zaccaria
- Department of Cell & Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.
| | | | | | | |
Collapse
|
35
|
Allen-Brady K, Robison R, Cannon D, Varvil T, Villalobos M, Pingree C, Leppert MF, Miller J, McMahon WM, Coon H. Genome-wide linkage in Utah autism pedigrees. Mol Psychiatry 2010; 15:1006-15. [PMID: 19455147 PMCID: PMC4023913 DOI: 10.1038/mp.2009.42] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 03/25/2009] [Accepted: 04/13/2009] [Indexed: 01/04/2023]
Abstract
Genetic studies of autism over the past decade suggest a complex landscape of multiple genes. In the face of this heterogeneity, studies that include large extended pedigrees may offer valuable insights, as the relatively few susceptibility genes within single large families may be more easily discerned. This genome-wide screen of 70 families includes 20 large extended pedigrees of 6-9 generations, 6 moderate-sized families of 4-5 generations and 44 smaller families of 2-3 generations. The Center for Inherited Disease Research (CIDR) provided genotyping using the Illumina Linkage Panel 12, a 6K single-nucleotide polymorphism (SNP) platform. Results from 192 subjects with an autism spectrum disorder (ASD) and 461 of their relatives revealed genome-wide significance on chromosome 15q, with three possibly distinct peaks: 15q13.1-q14 (heterogeneity LOD (HLOD)=4.09 at 29 459 872 bp); 15q14-q21.1 (HLOD=3.59 at 36 837 208 bp); and 15q21.1-q22.2 (HLOD=5.31 at 55 629 733 bp). Two of these peaks replicate earlier findings. There were additional suggestive results on chromosomes 2p25.3-p24.1 (HLOD=1.87), 7q31.31-q32.3 (HLOD=1.97) and 13q12.11-q12.3 (HLOD=1.93). Affected subjects in families supporting the linkage peaks found in this study did not reveal strong evidence for distinct phenotypic subgroups.
Collapse
Affiliation(s)
- K Allen-Brady
- Utah Autism Research Project, Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Maestrini E, Pagnamenta AT, Lamb JA, Bacchelli E, Sykes NH, Sousa I, Toma C, Barnby G, Butler H, Winchester L, Scerri TS, Minopoli F, Reichert J, Cai G, Buxbaum JD, Korvatska O, Schellenberg GD, Dawson G, Bildt AD, Minderaa RB, Mulder EJ, Morris AP, Bailey AJ, Monaco AP. High-density SNP association study and copy number variation analysis of the AUTS1 and AUTS5 loci implicate the IMMP2L-DOCK4 gene region in autism susceptibility. Mol Psychiatry 2010; 15:954-68. [PMID: 19401682 PMCID: PMC2934739 DOI: 10.1038/mp.2009.34] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Revised: 02/19/2009] [Accepted: 04/02/2009] [Indexed: 01/02/2023]
Abstract
Autism spectrum disorders are a group of highly heritable neurodevelopmental disorders with a complex genetic etiology. The International Molecular Genetic Study of Autism Consortium previously identified linkage loci on chromosomes 7 and 2, termed AUTS1 and AUTS5, respectively. In this study, we performed a high-density association analysis in AUTS1 and AUTS5, testing more than 3000 single nucleotide polymorphisms (SNPs) in all known genes in each region, as well as SNPs in non-genic highly conserved sequences. SNP genotype data were also used to investigate copy number variation within these regions. The study sample consisted of 127 and 126 families, showing linkage to the AUTS1 and AUTS5 regions, respectively, and 188 gender-matched controls. Further investigation of the strongest association results was conducted in an independent European family sample containing 390 affected individuals. Association and copy number variant analysis highlighted several genes that warrant further investigation, including IMMP2L and DOCK4 on chromosome 7. Evidence for the involvement of DOCK4 in autism susceptibility was supported by independent replication of association at rs2217262 and the finding of a deletion segregating in a sib-pair family.
Collapse
Affiliation(s)
- E Maestrini
- Department of Biology, University of Bologna, Bologna, Italy
| | - A T Pagnamenta
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - J A Lamb
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Centre for Integrated Genomic Medical Research, University of Manchester, Manchester, UK
| | - E Bacchelli
- Department of Biology, University of Bologna, Bologna, Italy
| | - N H Sykes
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - I Sousa
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - C Toma
- Department of Biology, University of Bologna, Bologna, Italy
| | - G Barnby
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - H Butler
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - L Winchester
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - T S Scerri
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - F Minopoli
- Department of Biology, University of Bologna, Bologna, Italy
| | - J Reichert
- Department of Psychiatry, Seaver Autism Research Center, Mount Sinai School of Medicine, New York, NY, USA
| | - G Cai
- Department of Psychiatry, Seaver Autism Research Center, Mount Sinai School of Medicine, New York, NY, USA
| | - J D Buxbaum
- Department of Psychiatry, Seaver Autism Research Center, Mount Sinai School of Medicine, New York, NY, USA
| | - O Korvatska
- Geriatric Research Education and Clinical Centre, Veterans Affairs Puget Sound Health Care System, Seattle Division, Seattle, WA, USA
| | - G D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - G Dawson
- Autism Speaks, New York, NY, USA
- Department of Psychology, University of Washington, Seattle, WA, USA
| | - A de Bildt
- Department of Psychiatry, Child and Adolescent Psychiatry, University Medical Center Groningen, Groningen, The Netherlands
| | - R B Minderaa
- Department of Psychiatry, Child and Adolescent Psychiatry, University Medical Center Groningen, Groningen, The Netherlands
| | - E J Mulder
- Department of Psychiatry, Child and Adolescent Psychiatry, University Medical Center Groningen, Groningen, The Netherlands
| | - A P Morris
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - A J Bailey
- University Department of Psychiatry, Warneford Hospital, Oxford, UK
| | - A P Monaco
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - IMGSAC12
- Department of Biology, University of Bologna, Bologna, Italy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Centre for Integrated Genomic Medical Research, University of Manchester, Manchester, UK
- Department of Psychiatry, Seaver Autism Research Center, Mount Sinai School of Medicine, New York, NY, USA
- Geriatric Research Education and Clinical Centre, Veterans Affairs Puget Sound Health Care System, Seattle Division, Seattle, WA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Autism Speaks, New York, NY, USA
- Department of Psychology, University of Washington, Seattle, WA, USA
- Department of Psychiatry, Child and Adolescent Psychiatry, University Medical Center Groningen, Groningen, The Netherlands
- University Department of Psychiatry, Warneford Hospital, Oxford, UK
| |
Collapse
|
37
|
Rucker JJH, McGuffin P. Polygenic heterogeneity: a complex model of genetic inheritance in psychiatric disorders. Biol Psychiatry 2010; 68:312-3. [PMID: 20674601 DOI: 10.1016/j.biopsych.2010.06.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Accepted: 06/28/2010] [Indexed: 10/19/2022]
Affiliation(s)
- James J H Rucker
- Medical Research Council, Social Genetic and Developmental Psychiatry Centre, The Institute of Psychiatry, King's College London, London, United Kingdom.
| | | |
Collapse
|
38
|
Pagnamenta AT, Bacchelli E, de Jonge MV, Mirza G, Scerri TS, Minopoli F, Chiocchetti A, Ludwig KU, Hoffmann P, Paracchini S, Lowy E, Harold DH, Chapman JA, Klauck SM, Poustka F, Houben RH, Staal WG, Ophoff RA, O'Donovan MC, Williams J, Nöthen MM, Schulte-Körne G, Deloukas P, Ragoussis J, Bailey AJ, Maestrini E, Monaco AP. Characterization of a family with rare deletions in CNTNAP5 and DOCK4 suggests novel risk loci for autism and dyslexia. Biol Psychiatry 2010; 68:320-8. [PMID: 20346443 PMCID: PMC2941017 DOI: 10.1016/j.biopsych.2010.02.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 01/15/2010] [Accepted: 02/01/2010] [Indexed: 11/16/2022]
Abstract
BACKGROUND Autism spectrum disorders (ASDs) are characterized by social, communication, and behavioral deficits and complex genetic etiology. A recent study of 517 ASD families implicated DOCK4 by single nucleotide polymorphism (SNP) association and a microdeletion in an affected sibling pair. METHODS The DOCK4 microdeletion on 7q31.1 was further characterized in this family using QuantiSNP analysis of 1M SNP array data and reverse transcription polymerase chain reaction. Extended family members were tested by polymerase chain reaction amplification of junction fragments. DOCK4 dosage was measured in additional samples using SNP arrays. Since QuantiSNP analysis identified a novel CNTNAP5 microdeletion in the same affected sibling pair, this gene was sequenced in 143 additional ASD families. Further polymerase chain reaction-restriction fragment length polymorphism analysis included 380 ASD cases and suitable control subjects. RESULTS The maternally inherited microdeletion encompassed chr7:110,663,978-111,257,682 and led to a DOCK4-IMMP2L fusion transcript. It was also detected in five extended family members with no ASD. However, six of nine individuals with this microdeletion had poor reading ability, which prompted us to screen 606 other dyslexia cases. This led to the identification of a second DOCK4 microdeletion co-segregating with dyslexia. Assessment of genomic background in the original ASD family detected a paternal 2q14.3 microdeletion disrupting CNTNAP5 that was also transmitted to both affected siblings. Analysis of other ASD cohorts revealed four additional rare missense changes in CNTNAP5. No exonic deletions of DOCK4 or CNTNAP5 were seen in 2091 control subjects. CONCLUSIONS This study highlights two new risk factors for ASD and dyslexia and demonstrates the importance of performing a high-resolution assessment of genomic background, even after detection of a rare and likely damaging microdeletion using a targeted approach.
Collapse
Affiliation(s)
- Alistair T. Pagnamenta
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Elena Bacchelli
- Department of Biology, University of Bologna, Bologna, Italy
| | - Maretha V. de Jonge
- Department of Child and Adolescent Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ghazala Mirza
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Thomas S. Scerri
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Andreas Chiocchetti
- Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Kerstin U. Ludwig
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Per Hoffmann
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Silvia Paracchini
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ernesto Lowy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Denise H. Harold
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom
| | - Jade A. Chapman
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom
| | - Sabine M. Klauck
- Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Fritz Poustka
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe-University, Frankfurt/Main, Germany
| | - Renske H. Houben
- Department of Child and Adolescent Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wouter G. Staal
- Department of Child and Adolescent Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roel A. Ophoff
- Department of Medical Genetics and Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
- University of California Los Angeles Center for Neurobehavioral Genetics, Los Angeles, California
| | | | - Julie Williams
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom
| | - Markus M. Nöthen
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Gerd Schulte-Körne
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Panos Deloukas
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Jiannis Ragoussis
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Anthony J. Bailey
- University Department of Psychiatry, Warneford Hospital, Oxford, United Kingdom
| | - Elena Maestrini
- Department of Biology, University of Bologna, Bologna, Italy
| | - Anthony P. Monaco
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
39
|
Williams EL, Casanova MF. Potential teratogenic effects of ultrasound on corticogenesis: implications for autism. Med Hypotheses 2010; 75:53-58. [PMID: 20149552 DOI: 10.1016/j.mehy.2010.01.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 01/20/2010] [Indexed: 02/03/2023]
Abstract
The phenotypic expression of autism, according to the Triple Hit Hypothesis, is determined by three factors: a developmental time window of vulnerability, genetic susceptibility, and environmental stressors. In utero exposure to thalidomide, valproic acid, and maternal infections are examples of some of the teratogenic agents which increase the risk of developing autism and define a time window of vulnerability. An additional stressor to genetically susceptible individuals during this time window of vulnerability may be prenatal ultrasound. Ultrasound enhances the genesis and differentiation of progenitor cells by activating the nitric oxide (NO) pathway and related neurotrophins. The effects of this pathway activation, however, are determined by the stage of development of the target cells, local concentrations of NO, and the position of nuclei (basal versus apical), causing consequent proliferation at some stages while driving differentiation and migration at others. Ill-timed activation or overactivation of this pathway by ultrasound may extend proliferation, increasing total cell number, and/or may trigger precipitous migration, causing maldistribution of neurons amongst cortical lamina, ganglia, white matter, and germinal zones. The rising rates of autism coincident with the increased use of ultrasound in obstetrics and its teratogenic/toxic effects on the CNS demand further research regarding a putative correlation.
Collapse
Affiliation(s)
- E L Williams
- Department of Psychiatry and Behavioral Sciences, University of Louisville, Louisville, KY, USA
| | | |
Collapse
|
40
|
Panaitof SC, Abrahams BS, Dong H, Geschwind DH, White SA. Language-related Cntnap2 gene is differentially expressed in sexually dimorphic song nuclei essential for vocal learning in songbirds. J Comp Neurol 2010; 518:1995-2018. [PMID: 20394055 PMCID: PMC2864722 DOI: 10.1002/cne.22318] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Multiple studies, involving distinct clinical populations, implicate contactin associated protein-like 2 (CNTNAP2) in aspects of language development and performance. While CNTNAP2 is broadly distributed in developing rodent brain, it shows a striking gradient of frontal cortical enrichment in developing human brain, consistent with a role in patterning circuits that subserve higher cognition and language. To test the hypothesis that CNTNAP2 may be important for learned vocal communication in additional species, we employed in situ hybridization to characterize transcript distribution in the zebra finch, an experimentally tractable songbird for which the neural substrate of this behavior is well established. Consistent with an important role in learned vocalization, Cntnap2 was enriched or diminished in key song control nuclei relative to adjacent brain tissue. Importantly, this punctuated expression was observed in males, but not females, in accord with the sexual dimorphism of neural circuitry and vocal learning in this species. Ongoing functional work will provide important insights into the relationship between Cntnap2 and vocal communication in songbirds and thereby clarify mechanisms at play in disorders of human cognition and language.
Collapse
Affiliation(s)
- S. Carmen Panaitof
- Department of Physiological Science, University of California, Los Angeles, CA, 90095
| | - Brett S. Abrahams
- Program in Neurobehavioral Genetics and Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095
| | - Hongmei Dong
- Program in Neurobehavioral Genetics and Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095
| | - Daniel H. Geschwind
- Program in Neurobehavioral Genetics and Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095
| | - Stephanie A. White
- Department of Physiological Science, University of California, Los Angeles, CA, 90095
| |
Collapse
|
41
|
Coon H, Villalobos ME, Robison RJ, Camp NJ, Cannon DS, Allen-Brady K, Miller JS, McMahon WM. Genome-wide linkage using the Social Responsiveness Scale in Utah autism pedigrees. Mol Autism 2010; 1:8. [PMID: 20678250 PMCID: PMC2913945 DOI: 10.1186/2040-2392-1-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 04/08/2010] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Autism Spectrum Disorders (ASD) are phenotypically heterogeneous, characterized by impairments in the development of communication and social behaviour and the presence of repetitive behaviour and restricted interests. Dissecting the genetic complexity of ASD may require phenotypic data reflecting more detail than is offered by a categorical clinical diagnosis. Such data are available from the Social Responsiveness Scale (SRS) which is a continuous, quantitative measure of social ability giving scores that range from significant impairment to above average ability. METHODS We present genome-wide results for 64 multiplex and extended families ranging from two to nine generations. SRS scores were available from 518 genotyped pedigree subjects, including affected and unaffected relatives. Genotypes from the Illumina 6 k single nucleotide polymorphism panel were provided by the Center for Inherited Disease Research. Quantitative and qualitative analyses were done using MCLINK, a software package that uses Markov chain Monte Carlo (MCMC) methods to perform multilocus linkage analysis on large extended pedigrees. RESULTS When analysed as a qualitative trait, linkage occurred in the same locations as in our previous affected-only genome scan of these families, with findings on chromosomes 7q31.1-q32.3 [heterogeneity logarithm of the odds (HLOD) = 2.91], 15q13.3 (HLOD = 3.64), and 13q12.3 (HLOD = 2.23). Additional positive qualitative results were seen on chromosomes 6 and 10 in regions that may be of interest for other neuropsychiatric disorders. When analysed as a quantitative trait, results replicated a peak found in an independent sample using quantitative SRS scores on chromosome 11p15.1-p15.4 (HLOD = 2.77). Additional positive quantitative results were seen on chromosomes 7, 9, and 19. CONCLUSIONS The SRS linkage peaks reported here substantially overlap with peaks found in our previous affected-only genome scan of clinical diagnosis. In addition, we replicated a previous SRS peak in an independent sample. These results suggest the SRS is a robust and useful phenotype measure for genetic linkage studies of ASD. Finally, analyses of SRS scores revealed linkage peaks overlapping with evidence from other studies of neuropsychiatric diseases. The information available from the SRS itself may, therefore, reveal locations for autism susceptibility genes that would not otherwise be detected.
Collapse
Affiliation(s)
- Hilary Coon
- Utah Autism Research Project, Department of Psychiatry and Division of Genetic Epidemiology, University of Utah, 650 Komas Drive, Suite 206, Salt Lake City, UT 84108, USA
| | - Michele E Villalobos
- Utah Autism Research Project, Department of Psychiatry and Division of Genetic Epidemiology, University of Utah, 650 Komas Drive, Suite 206, Salt Lake City, UT 84108, USA
| | - Reid J Robison
- Utah Autism Research Project, Department of Psychiatry and Division of Genetic Epidemiology, University of Utah, 650 Komas Drive, Suite 206, Salt Lake City, UT 84108, USA
| | - Nicola J Camp
- Utah Autism Research Project, Department of Psychiatry and Division of Genetic Epidemiology, University of Utah, 650 Komas Drive, Suite 206, Salt Lake City, UT 84108, USA
| | - Dale S Cannon
- Utah Autism Research Project, Department of Psychiatry and Division of Genetic Epidemiology, University of Utah, 650 Komas Drive, Suite 206, Salt Lake City, UT 84108, USA
| | - Kristina Allen-Brady
- Utah Autism Research Project, Department of Psychiatry and Division of Genetic Epidemiology, University of Utah, 650 Komas Drive, Suite 206, Salt Lake City, UT 84108, USA
| | - Judith S Miller
- Utah Autism Research Project, Department of Psychiatry and Division of Genetic Epidemiology, University of Utah, 650 Komas Drive, Suite 206, Salt Lake City, UT 84108, USA
| | - William M McMahon
- Utah Autism Research Project, Department of Psychiatry and Division of Genetic Epidemiology, University of Utah, 650 Komas Drive, Suite 206, Salt Lake City, UT 84108, USA
| |
Collapse
|
42
|
Sousa I, Clark TG, Holt R, Pagnamenta AT, Mulder EJ, Minderaa RB, Bailey AJ, Battaglia A, Klauck SM, Poustka F, Monaco AP. Polymorphisms in leucine-rich repeat genes are associated with autism spectrum disorder susceptibility in populations of European ancestry. Mol Autism 2010; 1:7. [PMID: 20678249 PMCID: PMC2913944 DOI: 10.1186/2040-2392-1-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 03/25/2010] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Autism spectrum disorders (ASDs) are a group of highly heritable neurodevelopmental disorders which are characteristically comprised of impairments in social interaction, communication and restricted interests/behaviours. Several cell adhesion transmembrane leucine-rich repeat (LRR) proteins are highly expressed in the nervous system and are thought to be key regulators of its development. Here we present an association study analysing the roles of four promising candidate genes - LRRTM1 (2p), LRRTM3 (10q), LRRN1 (3p) and LRRN3 (7q) - in order to identify common genetic risk factors underlying ASDs. METHODS In order to gain a better understanding of how the genetic variation within these four gene regions may influence susceptibility to ASDs, a family-based association study was undertaken in 661 families of European ancestry selected from four different ASD cohorts. In addition, a case-control study was undertaken across the four LRR genes, using logistic regression in probands with ASD of each population against 295 ECACC controls. RESULTS Significant results were found for LRRN3 and LRRTM3 (P < 0.005), using both single locus and haplotype approaches. These results were further supported by a case-control analysis, which also highlighted additional SNPs in LRRTM3. CONCLUSIONS Overall, our findings implicate the neuronal leucine-rich genes LRRN3 and LRRTM3 in ASD susceptibility.
Collapse
Affiliation(s)
- Inês Sousa
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sen B, Surindro Singh A, Sinha S, Chatterjee A, Ahmed S, Ghosh S, Usha R. Family-based studies indicate association ofEngrailed 2gene with autism in an Indian population. GENES BRAIN AND BEHAVIOR 2010; 9:248-55. [DOI: 10.1111/j.1601-183x.2009.00556.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Banaschewski T, Becker K, Scherag S, Franke B, Coghill D. Molecular genetics of attention-deficit/hyperactivity disorder: an overview. Eur Child Adolesc Psychiatry 2010; 19:237-57. [PMID: 20145962 PMCID: PMC2839490 DOI: 10.1007/s00787-010-0090-z] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 01/07/2010] [Indexed: 12/15/2022]
Abstract
As heritability is high in attention-deficit/hyperactivity disorder (ADHD), genetic factors must play a significant role in the development and course of this disorder. In recent years a large number of studies on different candidate genes for ADHD have been published, most have focused on genes involved in the dopaminergic neurotransmission system, such as DRD4, DRD5, DAT1/SLC6A3, DBH, DDC. Genes associated with the noradrenergic (such as NET1/SLC6A2, ADRA2A, ADRA2C) and serotonergic systems (such as 5-HTT/SLC6A4, HTR1B, HTR2A, TPH2) have also received considerable interest. Additional candidate genes related to neurotransmission and neuronal plasticity that have been studied less intensively include SNAP25, CHRNA4, NMDA, BDNF, NGF, NTF3, NTF4/5, GDNF. This review article provides an overview of these candidate gene studies, and summarizes findings from recently published genome-wide association studies (GWAS). GWAS is a relatively new tool that enables the identification of new ADHD genes in a hypothesis-free manner. Although these latter studies could be improved and need to be replicated they are starting to implicate processes like neuronal migration and cell adhesion and cell division as potentially important in the aetiology of ADHD and have suggested several new directions for future ADHD genetics studies.
Collapse
Affiliation(s)
- Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Mannheim, Germany.
| | | | | | | | | |
Collapse
|
45
|
Rommelse NNJ, Franke B, Geurts HM, Hartman CA, Buitelaar JK. Shared heritability of attention-deficit/hyperactivity disorder and autism spectrum disorder. Eur Child Adolesc Psychiatry 2010; 19:281-295. [PMID: 20148275 PMCID: PMC2839489 DOI: 10.1007/s00787-010-0092-x] [Citation(s) in RCA: 352] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Accepted: 01/08/2010] [Indexed: 01/17/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) are both highly heritable neurodevelopmental disorders. Evidence indicates both disorders co-occur with a high frequency, in 20-50% of children with ADHD meeting criteria for ASD and in 30-80% of ASD children meeting criteria for ADHD. This review will provide an overview on all available studies [family based, twin, candidate gene, linkage, and genome wide association (GWA) studies] shedding light on the role of shared genetic underpinnings of ADHD and ASD. It is concluded that family and twin studies do provide support for the hypothesis that ADHD and ASD originate from partly similar familial/genetic factors. Only a few candidate gene studies, linkage studies and GWA studies have specifically addressed this co-occurrence, pinpointing to some promising pleiotropic genes, loci and single nucleotide polymorphisms (SNPs), but the research field is in urgent need for better designed and powered studies to tackle this complex issue. We propose that future studies examining shared familial etiological factors for ADHD and ASD use a family-based design in which the same phenotypic (ADHD and ASD), candidate endophenotypic, and environmental measurements are obtained from all family members. Multivariate multi-level models are probably best suited for the statistical analysis.
Collapse
Affiliation(s)
- Nanda N J Rommelse
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Center, Reinier Postlaan 10, Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
46
|
Ozgen HM, Hop JW, Hox JJ, Beemer FA, van Engeland H. Minor physical anomalies in autism: a meta-analysis. Mol Psychiatry 2010; 15:300-7. [PMID: 18626481 DOI: 10.1038/mp.2008.75] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Autism is a complex neurodevelopmental disorder in which the interactions of genetic, epigenetic and environmental influences play a causal role. Despite the compelling evidence for a strong heritability, the etiology and molecular mechanisms underlying autism remain unclear. High phenotypic variability and genetic heterogeneity confounds the identification of susceptibility genes. The lack of robust indicators to tackle this complexity in autism has led researchers to seek for novel diagnostic tools to create homogenous subgroups. Several studies have indicated that patients with autism have higher rates of minor physical anomalies (MPAs) and that MPAs may serve as a diagnostic tool; however, the results have been inconsistent. Using the cumulative data from seven studies on MPAs in autism, this meta-analysis seeks to examine whether the aggregate data provide evidence of a large mean effect size and statistical significance for MPAs in autism. It covers the studies using multiple research methods till June 2007. The current results from seven studies suggested a significant association of MPAs in autism with a robust pooled effect size (d=0.84), and thereby provide the strongest evidence to date about the close association between MPAs and autism. Our results emphasize the importance of MPAs in the identification of heterogeneity in autism and suggest that the success of future autism genetics research will be exploited by the use of MPAs. Implications for the design of future studies on MPAs in autism are discussed and suggestions for further investigation of these important markers are proposed. Clarifying this relation might improve understanding of risk factors and molecular mechanisms in autism.
Collapse
Affiliation(s)
- H M Ozgen
- Department of Child and Adolescent Psychiatry, University Medical Centre, Utrecht, The Netherlands.
| | | | | | | | | |
Collapse
|
47
|
Genetics of autistic disorders: review and clinical implications. Eur Child Adolesc Psychiatry 2010; 19:169-78. [PMID: 19941018 PMCID: PMC2839494 DOI: 10.1007/s00787-009-0076-x] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 10/30/2009] [Indexed: 12/21/2022]
Abstract
Twin and family studies in autistic disorders (AD) have elucidated a high heritability of AD. In this literature review, we will present an overview on molecular genetic studies in AD and highlight the most recent findings of an increased rate of copy number variations in AD. An extensive literature search in the PubMed database was performed to obtain English published articles on genetic findings in autism. Results of linkage, (genome wide) association and cytogenetic studies are presented, and putative aetiopathological pathways are discussed. Implications of the different genetic findings for genetic counselling and genetic testing at present will be described. The article ends with a prospectus on future directions.
Collapse
|
48
|
Abstract
Cardiovascular disease, metabolic syndrome, schizophrenia, diabetes, bipolar disorder, and autism are a few of the numerous complex diseases for which researchers are trying to decipher the genetic composition. One interest of geneticists is to determine the quantitative trait loci (QTLs) that underlie the genetic portion of these diseases and their risk factors. The difficulty for researchers is that the QTLs underlying these diseases are likely to have small to medium effects which will necessitate having large studies in order to have adequate power. Combining information across multiple studies provides a way for researchers to potentially increase power while making the most of existing studies.Here, we will explore some of the methods that are currently being used by geneticists to combine information across multiple genome-wide linkage studies. There are two main types of meta-analyses: (1) those that yield a measure of significance, such as Fisher's p-value method along with its extensions/modifications and the genome search meta-analysis (GSMA) method, and (2) those that yield a measure of a common effect size and the corresponding standard error, such as model-based methods and Bayesian methods. Some of these methods allow for the assessment of heterogeneity. This chapter will conclude with a recommendation for usage.
Collapse
Affiliation(s)
- Trecia A Kippola
- Department of Statistics, Oklahoma State University, OK, Stillwater, USA
| | | |
Collapse
|
49
|
Jackson PB, Boccuto L, Skinner C, Collins JS, Neri G, Gurrieri F, Schwartz CE. Further evidence that the rs1858830 C variant in the promoter region of the MET gene is associated with autistic disorder. Autism Res 2009; 2:232-6. [PMID: 19681062 DOI: 10.1002/aur.87] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous studies in three independent cohorts have shown that the rs1858830 C allele variant in the promoter region of the MET gene on chromosome 7q31 is associated with autism. Another study has found correlations between other alterations in the MET gene and autism in two unrelated cohorts. This study screened two cohorts, an Autistic Disorder cohort from South Carolina and a Pervasive Developmental Disorder (PDD) cohort from Italy, for the presence of the C allele variant in rs1858830. A significant increase in the C allele variant frequency was found in the South Carolina Autistic Disorder patients as compared to South Carolina Controls (chi(2)=5.8, df=1, P=0.02). In the South Carolina cohort, a significant association with Autistic Disorder was found when comparing the CC and CG genotypes to the GG genotype (odds ratio (OR)=1.64; 95% confidence interval (CI)=1.12-2.40; chi(2)=6.5, df=1, P=0.01) in cases and controls. In the Italian cohort, no significant association with PDD was found when comparing the CC or CG genotype to the GG genotype (OR=1.20; 95% CI=0.56-2.56; chi(2)=0.2, df=1, P=0.64). This study is the third independent study to find the rs1858830 C variant in the MET gene promoter to be associated with autism.
Collapse
Affiliation(s)
- Pamela B Jackson
- JC Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, South Carolina, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Takahashi A, Tomihara K, Shiroishi T, Koide T. Genetic mapping of social interaction behavior in B6/MSM consomic mouse strains. Behav Genet 2009; 40:366-76. [PMID: 19936911 PMCID: PMC2853700 DOI: 10.1007/s10519-009-9312-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 11/02/2009] [Indexed: 11/27/2022]
Abstract
Genetic studies are indispensable for understanding the mechanisms by which individuals develop differences in social behavior. We report genetic mapping of social interaction behavior using inter-subspecific consomic strains established from MSM/Ms (MSM) and C57BL/6J (B6) mice. Two animals of the same strain and sex, aged 10 weeks, were introduced into a novel open-field for 10 min. Social contact was detected by an automated system when the distance between the centers of the two animals became less than ~12 cm. In addition, detailed behavioral observations were made of the males. The wild-derived mouse strain MSM showed significantly longer social contact as compared to B6. Analysis of the consomic panel identified two chromosomes (Chr 6 and Chr 17) with quantitative trait loci (QTL) responsible for lengthened social contact in MSM mice and two chromosomes (Chr 9 and Chr X) with QTL that inhibited social contact. Detailed behavioral analysis of males identified four additional chromosomes associated with social interaction behavior. B6 mice that contained Chr 13 from MSM showed more genital grooming and following than the parental B6 strain, whereas the presence of Chr 8 and Chr 12 from MSM resulted in a reduction of those behaviors. Longer social sniffing was observed in Chr 4 consomic strain than in B6 mice. Although the frequency was low, aggressive behavior was observed in a few pairs from consomic strains for Chrs 4, 13, 15 and 17, as well as from MSM. The social interaction test has been used as a model to measure anxiety, but genetic correlation analysis suggested that social interaction involves different aspects of anxiety than are measured by open-field test.
Collapse
Affiliation(s)
- Aki Takahashi
- Mouse Genomics Resource Laboratory, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540 Japan
- Department of Genetics, SOKENDAI, Hayama, Kanagawa, 240-0193 Japan
- Present Address: Department of Psychology, Tufts University, 530 Boston Avenue (Bacon Hall), Medford, MA 02155 USA
| | - Kazuya Tomihara
- Department of Psychology, Faculty of Law, Economics and Humanities, Kagoshima University, Kohrimoto, Kagoshima, 890-0065 Japan
| | - Toshihiko Shiroishi
- Mammalian Genetics Laboratory, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540 Japan
- Department of Genetics, SOKENDAI, Hayama, Kanagawa, 240-0193 Japan
| | - Tsuyoshi Koide
- Mouse Genomics Resource Laboratory, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540 Japan
- Department of Genetics, SOKENDAI, Hayama, Kanagawa, 240-0193 Japan
| |
Collapse
|