1
|
Chang X, Qu HQ, Liu Y, Glessner JT, Hakonarson H. Mitochondrial DNA Haplogroup K Is Protective Against Autism Spectrum Disorder Risk in Populations of European Ancestry. J Am Acad Child Adolesc Psychiatry 2024; 63:835-844. [PMID: 38072244 PMCID: PMC11186604 DOI: 10.1016/j.jaac.2023.09.550] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 09/23/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE Accumulative evidence indicates a critical role of mitochondrial function in autism spectrum disorders (ASD), implying that ASD risk may be linked to mitochondrial dysfunction due to DNA (mtDNA) variations. Although a few studies have explored the association between mtDNA variations and ASD, the role of mtDNA in ASD is still unclear. Here, we aimed to investigate whether mitochondrial DNA haplogroups are associated with the risk of ASD. METHOD Two European cohorts and an Ashkenazi Jewish (AJ) cohort were analyzed, including 2,062 ASD patients in comparison with 4,632 healthy controls. DNA samples were genotyped using Illumina HumanHap550/610 and Illumina 1M arrays, inclusive of mitochondrial markers. Mitochondrial DNA (mtDNA) haplogroups were identified from genotyping data using HaploGrep2. A mitochondrial genome imputation pipeline was established to detect mtDNA variants. We conducted a case-control study to investigate potential associations of mtDNA haplogroups and variants with the susceptibility to ASD. RESULTS We observed that the ancient adaptive mtDNA haplogroup K was significantly associated with decreased risk of ASD by the investigation of 2 European cohorts including a total of 2,006 cases and 4,435 controls (odds ratio = 0.64, P=1.79 × 10-5), and we replicated this association in an Ashkenazi Jewish (AJ) cohort including 56 cases and 197 controls (odds ratio = 0.35, P = 9.46 × 10-3). Moreover, we demonstrate that the mtDNA variants rs28358571, rs28358584, and rs28358280 are significantly associated with ASD risk. Further expression quantitative trait loci (eQTLs) analysis indicated that the rs28358584 and rs28358280 genotypes are associated with expression levels of nearby genes in brain tissues, suggesting those mtDNA variants may confer risk for ASD via regulation of expression levels of genes encoded by the mitochondrial genome. CONCLUSION This study helps to shed light on the contribution of mitochondria in ASD and provides new insights into the genetic mechanism underlying ASD, suggesting the potential involvement of mtDNA-encoded proteins in the development of ASD. PLAIN LANGUAGE SUMMARY Increasing evidence indicates that mitochondrial dysfunction may be linked to autism spectrum disorder (ASD). This study investigated potential associations of mitochondrial DNA (mtDNA) variants in 2 European and Ashkenazi Jewish cohorts including 2,062 individuals with ASD and 4,632 healthy controls. Researchers found that the ancient mtDNA haplogroup K was linked to a reduced risk of ASD in both European and Ashkenazi Jewish populations. Additionally, specific mtDNA variants were associated with ASD risk and were shown to influence the expression of nearby genes in the brain. These findings highlight the potential involvement of mtDNA in ASD development, offering new insights into the genetic mechanisms underlying the disorder.
Collapse
Affiliation(s)
- Xiao Chang
- Children's Hospital of Philadelphia, Pennsylvania, United States; Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China.
| | - Hui-Qi Qu
- Children's Hospital of Philadelphia, Pennsylvania, United States
| | - Yichuan Liu
- Children's Hospital of Philadelphia, Pennsylvania, United States
| | | | - Hakon Hakonarson
- Children's Hospital of Philadelphia, Pennsylvania, United States; The Perelman School of Medicine, University of Pennsylvania, Pennsylvania, United States and Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
2
|
Zhang Q, Wang Y, Tao J, Xia R, Zhang Y, Liu Z, Cheng J. Sex-biased single-cell genetic landscape in mice with autism spectrum disorder. J Genet Genomics 2024; 51:338-351. [PMID: 37703921 DOI: 10.1016/j.jgg.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/15/2023]
Abstract
Autistic spectrum disorder (ASD) is a male-biased, heterogeneous neurodevelopmental disorder that affects approximately 1%-2% of the population. Prenatal exposure to valproic acid (VPA) is a recognized risk factor for ASD, but the cellular and molecular basis of VPA-induced ASD at the single-cell resolution is unclear. Here, we aim to compare the cellular and molecular differences in the hippocampus between male and female prenatal mice with ASD at the single-cell transcriptomic level. The transcriptomes of more than 45,000 cells are assigned to 12 major cell types, including neurons, glial cells, vascular cells, and immune cells. Cell type-specific genes with altered expression after prenatal VPA exposure are analyzed, and the largest number of differentially expressed genes (DEGs) are found in neurons, choroid plexus epithelial cells, and microglia. In microglia, several pathways related to inflammation are found in both males and females, including the tumor necrosis factor (TNF), nuclear factor kappa B (NF-κB), toll-like receptor (TLR), and mitogen-activated protein kinase (MAPK) signaling pathways, which are important for the induction of autistic-like behavior. Additionally, we note that several X-linked genes, including Bex1, Bex3, and Gria3, were among the male-specific DEGs of neurons. This pioneering study describes the landscape of the transcriptome in the hippocampus of autistic mice. The elucidation of sexual differences could provide innovative strategies for the prevention and treatment of ASD.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Yongjie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jie Tao
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Ruixue Xia
- Department of Respiratory and Critical Care Medicine, Henan University Huaihe Hospital, Kaifeng, Henan 475099, China
| | - Yijie Zhang
- Department of Respiratory and Critical Care Medicine, Henan University Huaihe Hospital, Kaifeng, Henan 475099, China
| | - Zhirui Liu
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| | - Jiwei Cheng
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| |
Collapse
|
3
|
Liu J, Yan J, Qu F, Mo W, Yu H, Hu P, Zhang Z. A pilot study on glutamate receptor and carrier gene variants and risk of childhood autism spectrum. Metab Brain Dis 2023; 38:2477-2488. [PMID: 37578654 DOI: 10.1007/s11011-023-01272-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/26/2023] [Indexed: 08/15/2023]
Abstract
Imbalanced glutamate signaling has been implicated in the development of autism spectrum disorder (ASD). This case-control study was to examine single nucleotide polymorphisms (SNPs) in glutamate receptor and carrier genes and determine their association with childhood ASD in a Chinese Han population. A total of 12 SNPs in genes encoding glutamate receptors (GRM7 and GRM8) and carriers (SLC1A1 and SLC25A12) were examined in 249 autistic children and 353 healthy controls. The Childhood Autism Rating Scale (CARS) and its verbal communication domain were applied to evaluate the severity of the disease and language impairment, respectively. The T allele of rs2292813 in the SLC25A12 gene was significantly associated with an increased risk of ASD (odds ratio (OD) = 1.7, 95% confidence interval (CI): 1.1-2.6, P = 0.0107). Neither the genotypes nor allele distributions of other SNPs were associated with the risk of ASD. Notably, rs1800656 and rs2237731 in the GRM8 gene, but not other SNPs, were related to the severity of language impairment. All SNPs were not correlated with the overall severity of ASD. Our findings support associations between the SLC25A12 gene variant and the risk of childhood ASD, and between the GRM8 gene variant and the severity of language impairment in the Chinese Han population.
Collapse
Affiliation(s)
- Jun Liu
- Department of Clinical Laboratory, Affiliated Xiaoshan Hospital of Hangzhou Normal University, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311202, China.
| | - Jing Yan
- Department of Clinical Laboratory, Affiliated Xiaoshan Hospital of Hangzhou Normal University, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311202, China
| | - Fei Qu
- Department of Clinical Laboratory, Affiliated Xiaoshan Hospital of Hangzhou Normal University, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311202, China
| | - Weiming Mo
- Department of Clinical Laboratory, Affiliated Xiaoshan Hospital of Hangzhou Normal University, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311202, China
| | - Hong Yu
- Department of Clinical Psychology, Xiaoshan First Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Pingfang Hu
- Department of Clinical Laboratory, Affiliated Xiaoshan Hospital of Hangzhou Normal University, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311202, China
| | - Zengyu Zhang
- Department of Pediatrics, Xiaoshan First Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
4
|
Chalkiadaki K, Statoulla E, Zafeiri M, Haji N, Lacaille JC, Powell CM, Jafarnejad SM, Khoutorsky A, Gkogkas CG. Reversal of memory and autism-related phenotypes in Tsc2+/- mice via inhibition of Nlgn1. Front Cell Dev Biol 2023; 11:1205112. [PMID: 37293130 PMCID: PMC10244498 DOI: 10.3389/fcell.2023.1205112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/16/2023] [Indexed: 06/10/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare monogenic disorder co-diagnosed with high rates of autism and is caused by loss of function mutations in the TSC1 or TSC2 genes. A key pathway hyperactivated in TSC is the mammalian/mechanistic target of rapamycin complex 1 (mTORC1), which regulates cap-dependent mRNA translation. We previously demonstrated that exaggerated cap-dependent translation leads to autism-related phenotypes and increased mRNA translation and protein expression of Neuroligin 1 (Nlgn1) in mice. Inhibition of Nlgn1 expression reversed social behavior deficits in mice with increased cap-dependent translation. Herein, we report elevated translation of Nlgn1 mRNA and an increase in its protein expression. Genetic or pharmacological inhibition of Nlgn1 expression in Tsc2 +/- mice rescued impaired hippocampal mGluR-LTD, contextual discrimination and social behavior deficits in Tsc2 +/- mice, without correcting mTORC1 hyperactivation. Thus, we demonstrate that reduction of Nlgn1 expression in Tsc2 +/- mice is a new therapeutic strategy for TSC and potentially other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Kleanthi Chalkiadaki
- Biomedical Research Institute, Foundation for Research and Technology Hellas, Ioannina, Greece
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Elpida Statoulla
- Biomedical Research Institute, Foundation for Research and Technology Hellas, Ioannina, Greece
| | - Maria Zafeiri
- Biomedical Research Institute, Foundation for Research and Technology Hellas, Ioannina, Greece
| | - Nabila Haji
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA), Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, Montreal, QC, Canada
| | - Jean-Claude Lacaille
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA), Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, Montreal, QC, Canada
| | - Craig M. Powell
- Department of Neurobiology, Civitan International Research Center at UAB Heersink School of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, United States
| | - Seyed Mehdi Jafarnejad
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Arkady Khoutorsky
- Department of Anesthesia, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Christos G. Gkogkas
- Biomedical Research Institute, Foundation for Research and Technology Hellas, Ioannina, Greece
| |
Collapse
|
5
|
Glessner JT, Khan ME, Chang X, Liu Y, Otieno FG, Lemma M, Slaby I, Hain H, Mentch F, Li J, Kao C, Sleiman PMA, March ME, Connolly J, Hakonarson H. Rare recurrent copy number variations in metabotropic glutamate receptor interacting genes in children with neurodevelopmental disorders. J Neurodev Disord 2023; 15:14. [PMID: 37120522 PMCID: PMC10148449 DOI: 10.1186/s11689-023-09483-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/06/2023] [Indexed: 05/01/2023] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs), such as attention deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD), are examples of complex and partially overlapping phenotypes that often lack definitive corroborating genetic information. ADHD and ASD have complex genetic associations implicated by rare recurrent copy number variations (CNVs). Both of these NDDs have been shown to share similar biological etiologies as well as genetic pleiotropy. METHODS Platforms aimed at investigating genetic-based associations, such as high-density microarray technologies, have been groundbreaking techniques in the field of complex diseases, aimed at elucidating the underlying disease biology. Previous studies have uncovered CNVs associated with genes within shared candidate genomic networks, including glutamate receptor genes, across multiple different NDDs. To examine shared biological pathways across two of the most common NDDs, we investigated CNVs across 15,689 individuals with ADHD (n = 7920), ASD (n = 4318), or both (n = 3,416), as well as 19,993 controls. Cases and controls were matched by genotype array (i.e., Illumina array versions). Three case-control association studies each calculated and compared the observed vs. expected frequency of CNVs across individual genes, loci, pathways, and gene networks. Quality control measures of confidence in CNV-calling, prior to association analyses, included visual inspection of genotype and hybridization intensity. RESULTS Here, we report results from CNV analysis in search for individual genes, loci, pathways, and gene networks. To extend our previous observations implicating a key role of the metabotropic glutamate receptor (mGluR) network in both ADHD and autism, we exhaustively queried patients with ASD and/or ADHD for CNVs associated with the 273 genomic regions of interest within the mGluR gene network (genes with one or two degrees protein-protein interaction with mGluR 1-8 genes). Among CNVs in mGluR network genes, we uncovered CNTN4 deletions enriched in NDD cases (P = 3.22E - 26, OR = 2.49). Additionally, we uncovered PRLHR deletions in 40 ADHD cases and 12 controls (P = 5.26E - 13, OR = 8.45) as well as clinically diagnostic relevant 22q11.2 duplications and 16p11.2 duplications in 23 ADHD + ASD cases and 9 controls (P = 4.08E - 13, OR = 15.05) and 22q11.2 duplications in 34 ADHD + ASD cases and 51 controls (P = 9.21E - 9, OR = 3.93); those control samples were not with previous 22qDS diagnosis in their EHR records. CONCLUSION Together, these results suggest that disruption in neuronal cell-adhesion pathways confers significant risk to NDDs and showcase that rare recurrent CNVs in CNTN4, 22q11.2, and 16p11.2 are overrepresented in NDDs that constitute patients predominantly suffering from ADHD and ASD. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02286817 First Posted: 10 November 14, ClinicalTrials.gov Identifier: NCT02777931 first posted: 19 May 2016, ClinicalTrials.gov Identifier: NCT03006367 first posted: 30 December 2016, ClinicalTrials.gov Identifier: NCT02895906 first posted: 12 September 2016.
Collapse
Affiliation(s)
- Joseph T Glessner
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Munir E Khan
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Xiao Chang
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Yichuan Liu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - F George Otieno
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Maria Lemma
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Isabella Slaby
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Heather Hain
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Frank Mentch
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Jin Li
- Department of Cell Biology, the Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Charlly Kao
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Patrick M A Sleiman
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Michael E March
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - John Connolly
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
6
|
Tissue-wide cell-specific proteogenomic modeling reveals novel candidate risk genes in autism spectrum disorders. NPJ Syst Biol Appl 2022; 8:31. [PMID: 36068227 PMCID: PMC9448731 DOI: 10.1038/s41540-022-00243-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
Autism spectrum disorders (ASD) are a set of complex neurodevelopmental diseases characterized with repetitive behavioral patterns and communication disabilities. Using a systems biology method called MAPSD (Markov Affinity-based Proteogenomic Signal Diffusion) for joint modeling of proteome dynamics and a wide array of omics datasets, we identified a list of candidate ASD risk genes. Leveraging the collected biological signals as well as a large-scale protein-protein interaction network adjusted based on single cell resolution proteome properties in four brain regions, we observed an agreement between the known and the newly identified candidate genes that are spatially enriched in neuronal cells within cerebral cortex at the protein level. Moreover, we created a detailed subcellular localization enrichment map of the known and the identified genes across 32 micro-domains and showed that neuronal cells and neuropils share the largest fraction of signal enrichment in cerebral cortex. Notably, we showed that the identified genes are among the transcriptional biomarkers of inhibitory and excitatory neurons in human frontal cortex. Intersecting the identified genes with a single cell RNA-seq data on ASD brains further evidenced that 20 candidate genes, including GRIK1, EMX2, STXBP6, and KCNJ3 are disrupted in distinct cell-types. Moreover, we showed that ASD risk genes are predominantly distributed in certain human interactome modules, and that the identified genes may act as the regulator for some of the known ASD loci. In summary, our study demonstrated how tissue-wide cell-specific proteogenomic modeling can reveal candidate genes for brain disorders that can be supported by convergent lines of evidence.
Collapse
|
7
|
Su LD, Wang N, Han J, Shen Y. Group 1 Metabotropic Glutamate Receptors in Neurological and Psychiatric Diseases: Mechanisms and Prospective. Neuroscientist 2021; 28:453-468. [PMID: 34088252 PMCID: PMC9449437 DOI: 10.1177/10738584211021018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors
that are activated by glutamate in the central nervous system (CNS).
Basically, mGluRs contribute to fine-tuning of synaptic efficacy and
control the accuracy and sharpness of neurotransmission. Among eight
subtypes, mGluR1 and mGluR5 belong to group 1 (Gp1) family, and are
implicated in multiple CNS disorders, such as Alzheimer’s disease,
autism, Parkinson’s disease, and so on. In the present review, we
systematically discussed underlying mechanisms and prospective of Gp1
mGluRs in a group of neurological and psychiatric diseases, including
Alzheimer’s disease, Parkinson’s disease, autism spectrum disorder,
epilepsy, Huntington’s disease, intellectual disability, Down’s
syndrome, Rett syndrome, attention-deficit hyperactivity disorder,
addiction, anxiety, nociception, schizophrenia, and depression, in
order to provide more insights into the therapeutic potential of Gp1
mGluRs.
Collapse
Affiliation(s)
- Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Na Wang
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ying Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
Reduced visual cortical plasticity in autism spectrum disorder. Brain Res Bull 2021; 170:11-21. [PMID: 33545310 DOI: 10.1016/j.brainresbull.2021.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 11/22/2022]
Abstract
There is increasing evidence implicating altered NMDA-receptor function in autism spectrum disorder (ASD). To investigate potential alterations in NMDA-dependent cortical plasticity in ASD, we examined the effect of visual high-frequency stimulation (HFS) on changes in plasticity in the visual cortex, measured by persistent changes in visual evoked potentials (VEPs), in individuals with ASD (n = 16) and neurotypical controls (NT; n = 15). VEPs were elicited by a checkerboard circle (0.83 Hz, 2-min blocks) at baseline and at 2, 4, and 20 min following exposure to HFS (8.87 Hz, 2 min), previously shown to induce LTP-like changes in the visual cortex. Difference waves were created by subtracting VEPs measured at baseline from each Post-HFS measure, and group differences assessed. We found that HFS resulted in reduced short-term potentiation of VEPs in ASD compared to NT participants. Thus, whilst ASD participants showed significant potentiation of the VEP immediately after HFS, this enhancement was not maintained, and only persisted into the second post-HFS assessment block in NT participants. Notably, ASD individuals who self-reported being more sensitive to visual stimuli showed greater shorter-term potentiation following visual HFS. Critically, there were no group differences in degree of neural entrainment to the visual HFS, or in attentional vigilance and task performance. These findings suggests that visual cortical plasticity is atypical in ASD, results consistent with reported altered NMDA receptor function in ASD.
Collapse
|
9
|
Ashitha SNM, Ramachandra NB. Integrated Functional Analysis Implicates Syndromic and Rare Copy Number Variation Genes as Prominent Molecular Players in Pathogenesis of Autism Spectrum Disorders. Neuroscience 2020; 438:25-40. [DOI: 10.1016/j.neuroscience.2020.04.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 01/05/2023]
|
10
|
Lin BD, Colas F, Nijman IJ, Medic J, Brands W, Parr JR, van Eijk KR, Klauck SM, Chiocchetti AG, Freitag CM, Maestrini E, Bacchelli E, Coon H, Vicente A, Oliveira G, Pagnamenta AT, Gallagher L, Ennis S, Anney R, Bourgeron T, Luykx JJ, Vorstman J. The role of rare compound heterozygous events in autism spectrum disorder. Transl Psychiatry 2020; 10:204. [PMID: 32572023 PMCID: PMC7308334 DOI: 10.1038/s41398-020-00866-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 05/05/2020] [Accepted: 05/15/2020] [Indexed: 11/30/2022] Open
Abstract
The identification of genetic variants underlying autism spectrum disorders (ASDs) may contribute to a better understanding of their underlying biology. To examine the possible role of a specific type of compound heterozygosity in ASD, namely, the occurrence of a deletion together with a functional nucleotide variant on the remaining allele, we sequenced 550 genes in 149 individuals with ASD and their deletion-transmitting parents. This approach allowed us to identify additional sequence variants occurring in the remaining allele of the deletion. Our main goal was to compare the rate of sequence variants in remaining alleles of deleted regions between probands and the deletion-transmitting parents. We also examined the predicted functional effect of the identified variants using Combined Annotation-Dependent Depletion (CADD) scores. The single nucleotide variant-deletion co-occurrence was observed in 13.4% of probands, compared with 8.1% of parents. The cumulative burden of sequence variants (n = 68) in pooled proband sequences was higher than the burden in pooled sequences from the deletion-transmitting parents (n = 41, X2 = 6.69, p = 0.0097). After filtering for those variants predicted to be most deleterious, we observed 21 of such variants in probands versus 8 in their deletion-transmitting parents (X2 = 5.82, p = 0.016). Finally, cumulative CADD scores conferred by these variants were significantly higher in probands than in deletion-transmitting parents (burden test, β = 0.13; p = 1.0 × 10-5). Our findings suggest that the compound heterozygosity described in the current study may be one of several mechanisms explaining variable penetrance of CNVs with known pathogenicity for ASD.
Collapse
Affiliation(s)
- Bochao Danae Lin
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Fabrice Colas
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Isaac J Nijman
- Department of Medical Informatics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jelena Medic
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - William Brands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jeremy R Parr
- Institute of Neuroscience, Newcastle University, Newcastle, UK
| | - Kristel R van Eijk
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sabine M Klauck
- Division of Molecular Genome Analysis and Division of Cancer Genome Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas G Chiocchetti
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, JW Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Christine M Freitag
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, JW Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Elena Maestrini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Elena Bacchelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Hilary Coon
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Astrid Vicente
- Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, Lisboa, Portugal
| | | | - Alistair T Pagnamenta
- NIHR Oxford BRC, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Louise Gallagher
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity College Dublin, Trinity Centre for Health Sciences, Dublin, Ireland
| | - Sean Ennis
- Academic Centre on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Richard Anney
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR3571 CNRS, Université de Paris, Paris, France
| | - Jurjen J Luykx
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- GGNet Mental Health, Apeldoorn, The Netherlands
| | - Jacob Vorstman
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- Program in Genetics and Genome Biology, Research Institute, and Department of Psychiatry, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Light-Adapted Electroretinogram Differences in Autism Spectrum Disorder. J Autism Dev Disord 2020; 50:2874-2885. [DOI: 10.1007/s10803-020-04396-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
12
|
Larijani B, Foroughi Heravani N, Alavi-Moghadam S, Goodarzi P, Rezaei-Tavirani M, Payab M, Gholami M, Razi F, Arjmand B. Cell Therapy Targets for Autism Spectrum Disorders: Hopes, Challenges and Future Directions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1341:107-124. [PMID: 32072476 DOI: 10.1007/5584_2020_491] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Autism spectrum disorders as a group of pediatric neurodevelopmental diseases is a crucial part of the worldwide disabilities which have influence in communication skills, social interactions, and ability to understand the concepts. The precise pathophysiology of autism spectrum disorders due to the abundance of involved mechanisms is unknown. Some of these involved mechanisms are related to genetic factors, chronic neuro inflammation, mitochondrial dysfunction, oxidative stress, immune dysregulation, hormonal imbalance, and environmental factors. Current main treatments for autisms are behavioral, nutritional and medical therapies, however there is not definitive treatment approach. Therein, more novel therapies are still required to improve the symptoms. Several preclinical and clinical evidence were shown that stem cell therapy is a potential treatment option for autism spectrum disorders individuals. Considering the significant factors which can affect the outcome of stem cell therapeutic effects including stem cell types, route and dosage of administration, and mechanism of activity along with selecting best animal models can be very important in performing clinical trials.
Collapse
Affiliation(s)
- Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Foroughi Heravani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology & Pharmacology, Faculty of Pharmacy; Toxicology and Poisoning Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Razi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Siences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Chong CH, Li Q, Mak PHS, Ng CCP, Leung EHW, Tan VH, Chan AKW, McAlonan G, Chan SY. Lrrc7 mutant mice model developmental emotional dysregulation that can be alleviated by mGluR5 allosteric modulation. Transl Psychiatry 2019; 9:244. [PMID: 31582721 PMCID: PMC6776540 DOI: 10.1038/s41398-019-0580-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 01/30/2023] Open
Abstract
LRRC7 has been identified as a candidate gene for severe childhood emotional dysregulation. Direct experimental evidence for a role of LRRC7 in the disease is needed, as is a better understanding of its impact on neuronal structure and signaling, and hence potential treatment targets. Here, we generated and analyzed an Lrrc7 mutant mouse line. Consistent with a critical role of LRRC7 in emotional regulation, mutant mice had inappropriate juvenile aggressive behavior and significant anxiety-like behavior and social dysfunction in adulthood. The pivotal role of mGluR5 signaling was demonstrated by rescue of behavioral defects with augmentation of mGluR5 receptor activity by 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB). Intra-peritoneal injection of CDPPB alleviated abnormal juvenile behavior, as well as anxiety-like behavior and hypersociability at adulthood. Furthermore, mutant primary neurons had impaired neurite outgrowth which was rescued by CDPPB treatment. In conclusion, Lrrc7 mutant mice provide a valuable tool to model childhood emotional dysregulation and persistent mental health comorbidities. Moreover, our data highlight an important role of LRRC7 in mGluR5 signaling, which is a potential new treatment target for anxiety and social dysfunction.
Collapse
Affiliation(s)
- Chi Ho Chong
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qi Li
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Priscilla Hoi Shan Mak
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cypress Chun Pong Ng
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Eva Hin Wa Leung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Vicky Huiqi Tan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anthony Kin Wang Chan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Grainne McAlonan
- The Sackler Centre for Translational Neurodevelopment and The Department of Forensic and Neurodevelopmental Sciences, King's College London, The South London and Maudsley NHS Foundation Trust, London, UK
| | - Siu Yuen Chan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
14
|
Bellosta P, Soldano A. Dissecting the Genetics of Autism Spectrum Disorders: A Drosophila Perspective. Front Physiol 2019; 10:987. [PMID: 31481894 PMCID: PMC6709880 DOI: 10.3389/fphys.2019.00987] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 07/18/2019] [Indexed: 01/10/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex group of multi-factorial developmental disorders that leads to communication and behavioral defects. Genetic alterations have been identified in around 20% of ASD patients and the use of genetic models, such as Drosophila melanogaster, has been of paramount importance in deciphering the significance of these alterations. In fact, many of the ASD associated genes, such as FMR1, Neurexin, Neuroligins and SHANK encode for proteins that have conserved functions in neurons and during synapse development, both in humans and in the fruit fly. Drosophila is a prominent model in neuroscience due to the conserved genetic networks that control neurodevelopmental processes and to the ease of manipulating its genetics. In the present review we will describe recent advances in the field of ASD with a particular focus on the characterization of genes where the use of Drosophila has been fundamental to better understand their function.
Collapse
Affiliation(s)
- Paola Bellosta
- Laboratory of Metabolism of Cell Growth and Neuronal Survival, Department of Cellular, Computational and Integrative Biology (CIBio), University of Trento, Trento, Italy.,Department of Medicine, New York University Langone Medical Center, New York, NY, United States
| | - Alessia Soldano
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBio), University of Trento, Trento, Italy
| |
Collapse
|
15
|
Vicari S, Napoli E, Cordeddu V, Menghini D, Alesi V, Loddo S, Novelli A, Tartaglia M. Copy number variants in autism spectrum disorders. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:421-427. [PMID: 30797015 DOI: 10.1016/j.pnpbp.2019.02.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 11/24/2022]
Abstract
In recent years, there has been an explosive increase in genetic studies related to autism spectrum disorder (ASD). This implicated the accumulation of a large amount of molecular data that may be used to verify various hypotheses and models developed to explore the complex genetic component of ASD. Several lines of evidence support the view that structural genomic variation contributes to the pathogenesis of ASD. The introduction of more sophisticated techniques for whole-genome screening, including array comparative genome hybridization and high-resolution single nucleotide polymorphism analysis, has allowed to identify an increasing number of ASD susceptibility loci. Copy number variants (CNVs) are the most common type of structural variation in the human genome and are considered important contributors to the pathogenesis of neurodevelopmental disorders, including ASD. In this review, we describe the accumulated evidence concerning the genetic events associated with ASD, and summarize current knowledge about the clinical relevance of CNVs in these disorders.
Collapse
Affiliation(s)
- Stefano Vicari
- Department of Neuroscience, Child Neuropsychiatric Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Eleonora Napoli
- Department of Neuroscience, Child Neuropsychiatric Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Viviana Cordeddu
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Deny Menghini
- Department of Neuroscience, Child Neuropsychiatric Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Viola Alesi
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sara Loddo
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonio Novelli
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
16
|
Jin LE, Wang M, Galvin VC, Lightbourne TC, Conn PJ, Arnsten AFT, Paspalas CD. mGluR2 versus mGluR3 Metabotropic Glutamate Receptors in Primate Dorsolateral Prefrontal Cortex: Postsynaptic mGluR3 Strengthen Working Memory Networks. Cereb Cortex 2019; 28:974-987. [PMID: 28108498 DOI: 10.1093/cercor/bhx005] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Indexed: 01/06/2023] Open
Abstract
The newly evolved circuits in layer III of primate dorsolateral prefrontal cortex (dlPFC) generate the neural representations that subserve working memory. These circuits are weakened by increased cAMP-K+ channel signaling, and are a focus of pathology in schizophrenia, aging, and Alzheimer's disease. Cognitive deficits in these disorders are increasingly associated with insults to mGluR3 metabotropic glutamate receptors, while reductions in mGluR2 appear protective. This has been perplexing, as mGluR3 has been considered glial receptors, and mGluR2 and mGluR3 have been thought to have similar functions, reducing glutamate transmission. We have discovered that, in addition to their astrocytic expression, mGluR3 is concentrated postsynaptically in spine synapses of layer III dlPFC, positioned to strengthen connectivity by inhibiting postsynaptic cAMP-K+ channel actions. In contrast, mGluR2 is principally presynaptic as expected, with only a minor postsynaptic component. Functionally, increase in the endogenous mGluR3 agonist, N-acetylaspartylglutamate, markedly enhanced dlPFC Delay cell firing during a working memory task via inhibition of cAMP signaling, while the mGluR2 positive allosteric modulator, BINA, produced an inverted-U dose-response on dlPFC Delay cell firing and working memory performance. These data illuminate why insults to mGluR3 would erode cognitive abilities, and support mGluR3 as a novel therapeutic target for higher cognitive disorders.
Collapse
Affiliation(s)
- Lu E Jin
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Veronica C Galvin
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Taber C Lightbourne
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Peter Jeffrey Conn
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt UniversityMedical Center, Nashville, TN 37232-0697, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | | |
Collapse
|
17
|
Hoehe MR, Morris-Rosendahl DJ. The role of genetics and genomics in clinical psychiatry. DIALOGUES IN CLINICAL NEUROSCIENCE 2019. [PMID: 30581286 PMCID: PMC6296395 DOI: 10.31887/dcns.2018.20.3/mhoehe] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The enormous successes in the genetics and genomics of many diseases have provided the basis for the advancement of precision medicine. Thus, the detection of genetic variants associated with neuropsychiatric disorders, as well as treatment outcome, has raised growing expectations that these findings could soon be translated into the clinic to improve diagnosis, the prediction of disease risk and individual response to drug therapy. In this article, we will provide an introduction to the search for genes involved in psychiatric illness and summarize the present findings in major psychiatric disorders. We will review the genetic variants in genes encoding drug metabolizing enzymes and specific drug targets which were found to be associated with variable drug response and severe side effects. We will evaluate the clinical translatability of these findings, whether there is currently any role for genetic testing and in this context, make valuable sources of information available to the clinician seeking guidance and advice in this rapidly developing field of psychiatric genetics.
Collapse
Affiliation(s)
- Margret R Hoehe
- Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | - Deborah J Morris-Rosendahl
- Clinical Genetics and Genomics, Royal Brompton and Harefield NHS Foundation Trust, London, UK, NHLI, Imperial College London, UK
| |
Collapse
|
18
|
Fluorescent Light Incites a Conserved Immune and Inflammatory Genetic Response within Vertebrate Organs ( Danio Rerio, Oryzias Latipes and Mus Musculus). Genes (Basel) 2019; 10:genes10040271. [PMID: 30987199 PMCID: PMC6523474 DOI: 10.3390/genes10040271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 12/29/2022] Open
Abstract
Fluorescent light (FL) has been utilized for ≈60 years and has become a common artificial light source under which animals, including humans, spend increasing amounts of time. Although the solar spectrum is quite dissimilar in both wavelengths and intensities, the genetic consequences of FL exposure have not been investigated. Herein, we present comparative RNA-Seq results that establish expression patterns within skin, brain, and liver for Danio rerio, Oryzias latipes, and the hairless mouse (Mus musculus) after exposure to FL. These animals represent diurnal and nocturnal lifestyles, and ≈450 million years of evolutionary divergence. In all three organisms, FL induced transcriptional changes of the acute phase response signaling pathway and modulated inflammation and innate immune responses. Our pathway and gene clustering analyses suggest cellular perception of oxidative stress is promoting induction of primary up-stream regulators IL1B and TNF. The skin and brain of the three animals as well as the liver of both fish models all exhibit increased inflammation and immune responses; however, the mouse liver suppressed the same pathways. Overall, the conserved nature of the genetic responses observed after FL exposure, among fishes and a mammal, suggest the presence of light responsive genetic circuitry deeply embedded in the vertebrate genome.
Collapse
|
19
|
Kappel DB, Schuch JB, Rovaris DL, da Silva BS, Müller D, Breda V, Teche SP, S Riesgo R, Schüler-Faccini L, Rohde LA, Grevet EH, Bau CHD. ADGRL3 rs6551665 as a Common Vulnerability Factor Underlying Attention-Deficit/Hyperactivity Disorder and Autism Spectrum Disorder. Neuromolecular Med 2019; 21:60-67. [PMID: 30652248 DOI: 10.1007/s12017-019-08525-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 01/10/2019] [Indexed: 12/27/2022]
Abstract
Neurodevelopmental disorders are prevalent, frequently occur in comorbidity and share substantial genetic correlation. Previous evidence has suggested a role for the ADGRL3 gene in Attention-Deficit/Hyperactivity Disorder (ADHD) susceptibility in several samples. Considering ADGRL3 functionality in central nervous system development and its previous association with neurodevelopmental disorders, we aimed to assess ADGRL3 influence in early-onset ADHD (before 7 years of age) and Autism Spectrum Disorder (ASD). The sample comprises 187 men diagnosed with early-onset ADHD, 135 boys diagnosed with ASD and 468 male blood donors. We tested the association of an ADGRL3 variant (rs6551665) with both early-onset ADHD and ASD susceptibility. We observed significant associations between ADGRL3-rs6551665 on ADHD and ASD susceptibilities; we found that G-carriers were at increased risk of ADHD and ASD, in accordance with previous studies. The overall evidence from the literature, corroborated by our results, suggests that ADGRL3 might be involved in brain development, and genetic modifications related to it might be part of a shared vulnerability factor associated with the underlying neurobiology of neurodevelopmental disorders such as ADHD and ASD.
Collapse
Affiliation(s)
- Djenifer B Kappel
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil.,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Jaqueline B Schuch
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil.,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Graduate Program in Biomedical Gerontology, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Diego L Rovaris
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil.,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Bruna S da Silva
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil.,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Diana Müller
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil.,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Vitor Breda
- ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Stefania P Teche
- ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rudimar S Riesgo
- Child Neurology Unit, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lavínia Schüler-Faccini
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil
| | - Luís A Rohde
- ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,National Institute of Developmental Psychiatry for Children and Adolescents, Porto Alegre, Brazil
| | - Eugenio H Grevet
- ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Claiton H D Bau
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, UFRGS, Avenida Bento Gonçalves, 9500, Porto Alegre, RS, CEP: 91501-970, Brazil. .,ADHD Outpatient Program - Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
20
|
Brown EA, Lautz JD, Davis TR, Gniffke EP, VanSchoiack AAW, Neier SC, Tashbook N, Nicolini C, Fahnestock M, Schrum AG, Smith SEP. Clustering the autisms using glutamate synapse protein interaction networks from cortical and hippocampal tissue of seven mouse models. Mol Autism 2018; 9:48. [PMID: 30237867 PMCID: PMC6139139 DOI: 10.1186/s13229-018-0229-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022] Open
Abstract
Background Autism spectrum disorders (ASDs) are a heterogeneous group of behaviorally defined disorders and are associated with hundreds of rare genetic mutations and several environmental risk factors. Mouse models of specific risk factors have been successful in identifying molecular mechanisms associated with a given factor. However, comparisons among different models to elucidate underlying common pathways or to define clusters of biologically relevant disease subtypes have been complicated by different methodological approaches or different brain regions examined by the labs that developed each model. Here, we use a novel proteomic technique, quantitative multiplex co-immunoprecipitation or QMI, to make a series of identical measurements of a synaptic protein interaction network in seven different animal models. We aim to identify molecular disruptions that are common to multiple models. Methods QMI was performed on 92 hippocampal and cortical samples taken from seven mouse models of ASD: Shank3B, Shank3Δex4-9, Ube3a2xTG, TSC2, FMR1, and CNTNAP2 mutants, as well as E12.5 VPA (maternal valproic acid injection on day 12.5 post-conception). The QMI panel targeted a network of 16 interacting, ASD-linked, synaptic proteins, probing 240 potential co-associations. A custom non-parametric statistical test was used to call significant differences between ASD models and littermate controls, and Hierarchical Clustering by Principal Components was used to cluster the models using mean log2 fold change values. Results Each model displayed a unique set of disrupted interactions, but some interactions were disrupted in multiple models. These tended to be interactions that are known to change with synaptic activity. Clustering revealed potential relationships among models and suggested deficits in AKT signaling in Ube3a2xTG mice, which were confirmed by phospho-western blots. Conclusions These data highlight the great heterogeneity among models, but suggest that high-dimensional measures of a synaptic protein network may allow differentiation of subtypes of ASD with shared molecular pathology.
Collapse
Affiliation(s)
- Emily A Brown
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA
| | - Jonathan D Lautz
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA
| | - Tessa R Davis
- 2Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN USA.,3Present address: Department of Biomedical Engineering, UT Austin, Austin, TX USA
| | - Edward P Gniffke
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA
| | - Alison A W VanSchoiack
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA.,Present address: Nanostring, Seattle, WA USA
| | - Steven C Neier
- 2Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN USA.,5Present address: Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA USA.,6Present address: Department of Medicine, Harvard Medical School, Boston, MA USA.,7Present address: Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Noah Tashbook
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA
| | - Chiara Nicolini
- 8Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON Canada
| | - Margaret Fahnestock
- 8Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON Canada
| | - Adam G Schrum
- 9Departments of Molecular Microbiology & Immunology, Surgery and Bioengineering, University of Missouri, Columbia, MO USA
| | - Stephen E P Smith
- 1Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA USA.,10Department of Pediatrics and Graduate Program in Neuroscience, University of Washington, Seattle, WA USA
| |
Collapse
|
21
|
Hoehe MR. The role of genetics and genomics in clinical psychiatry. DIALOGUES IN CLINICAL NEUROSCIENCE 2018; 20:169-177. [PMID: 30581286 PMCID: PMC6296395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/15/2024]
Abstract
The enormous successes in the genetics and genomics of many diseases have provided the basis for the advancement of precision medicine. Thus, the detection of genetic variants associated with neuropsychiatric disorders, as well as treatment outcome, has raised growing expectations that these findings could soon be translated into the clinic to improve diagnosis, the prediction of disease risk and individual response to drug therapy. In this article, we will provide an introduction to the search for genes involved in psychiatric illness and summarize the present findings in major psychiatric disorders. We will review the genetic variants in genes encoding drug metabolizing enzymes and specific drug targets which were found to be associated with variable drug response and severe side effects. We will evaluate the clinical translatability of these findings, whether there is currently any role for genetic testing and in this context, make valuable sources of information available to the clinician seeking guidance and advice in this rapidly developing field of psychiatric genetics.
Collapse
|
22
|
Sharp BM. Basolateral amygdala, nicotinic cholinergic receptors, and nicotine: Pharmacological effects and addiction in animal models and humans. Eur J Neurosci 2018; 50:2247-2254. [PMID: 29802666 DOI: 10.1111/ejn.13970] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 01/08/2023]
Abstract
The amygdala is involved in processing incoming information about rewarding stimuli and emotions that denote danger such as anxiety and fear. Bi-directional neural connections between basolateral amygdala (BLA) and brain regions such as nucleus accumbens, prefrontal cortex, hippocampus, and hindbrain regions regulate motivation, cognition, and responses to stress. Altered local regulation of BLA excitability is pivotal to the behavioral disturbances characteristic of posttraumatic stress disorder, and relapse to drug use induced by stress. Herein, we review the physiological regulation of BLA by cholinergic inputs, emphasizing the role of BLA nicotinic receptors. We review BLA-dependent effects of nicotine on cognition, motivated behaviors, and emotional states, including memory, taking and seeking drugs, and anxiety and fear in humans and animal models. The alterations in BLA activity observed in animal studies inform human behavioral and brain imaging research by enabling a more exact understanding of altered BLA function. Converging evidence indicates that cholinergic signaling from basal forebrain projections to local nicotinic receptors is an important physiological regulator of BLA and that nicotine alters BLA function. In essence, BLA is necessary for behavioral responses to stimuli that evoke anxiety and fear; reinstatement of cue-induced drug seeking; responding to second-order cues conditioned to abused drugs; reacquisition of amplified nicotine self-administration due to chronic stress during abstinence; and to promote responding for natural reward.
Collapse
Affiliation(s)
- Burt M Sharp
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
23
|
Zamani Dadaneh S, Zhou M, Qian X. Covariate-dependent negative binomial factor analysis of RNA sequencing data. Bioinformatics 2018; 34:i61-i69. [PMID: 29949981 PMCID: PMC6022606 DOI: 10.1093/bioinformatics/bty237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Motivation High-throughput sequencing technologies, in particular RNA sequencing (RNA-seq), have become the basic practice for genomic studies in biomedical research. In addition to studying genes individually, for example, through differential expression analysis, investigating co-ordinated expression variations of genes may help reveal the underlying cellular mechanisms to derive better understanding and more effective prognosis and intervention strategies. Although there exists a variety of co-expression network based methods to analyze microarray data for this purpose, instead of blindly extending these methods for microarray data that may introduce unnecessary bias, it is crucial to develop methods well adapted to RNA-seq data to identify the functional modules of genes with similar expression patterns. Results We have developed a fully Bayesian covariate-dependent negative binomial factor analysis (dNBFA) method-dNBFA-for RNA-seq count data, to capture coordinated gene expression changes, while considering effects from covariates reflecting different influencing factors. Unlike existing co-expression network based methods, our proposed model does not require multiple ad-hoc choices on data processing, transformation, as well as co-expression measures and can be directly applied to RNA-seq data. Furthermore, being capable of incorporating covariate information, the proposed method can tackle setups with complex confounding factors in different experiment designs. Finally, the natural model parameterization removes the need for a normalization preprocessing step, as commonly adopted to compensate for the effect of sequencing-depth variations. Efficient Bayesian inference of model parameters is derived by exploiting conditional conjugacy via novel data augmentation techniques. Experimental results on several real-world RNA-seq datasets on complex diseases suggest dNBFA as a powerful tool for discovering the gene modules with significant differential expression and meaningful biological insight. Availability and implementation dNBFA is implemented in R language and is available at https://github.com/siamakz/dNBFA.
Collapse
Affiliation(s)
- Siamak Zamani Dadaneh
- Department of Electrical and Computer Engineering, TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, TX, USA
| | - Mingyuan Zhou
- Department of Information, Risk, and Operations Management, The University of Texas at Austin, Austin, TX, USA
| | - Xiaoning Qian
- Department of Electrical and Computer Engineering, TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, TX, USA
| |
Collapse
|
24
|
Shen L, Zhang K, Feng C, Chen Y, Li S, Iqbal J, Liao L, Zhao Y, Zhai J. iTRAQ-Based Proteomic Analysis Reveals Protein Profile in Plasma from Children with Autism. Proteomics Clin Appl 2018; 12:e1700085. [PMID: 29274201 DOI: 10.1002/prca.201700085] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 11/26/2017] [Indexed: 12/21/2022]
Abstract
PURPOSE Autism is a childhood neurological disorder with poorly understood etiology and pathology. This study is designed to identify differentially expressed proteins that might serve as potential biomarkers for autism. EXPERIMENTAL DESIGN We perform iTRAQ (isobaric tags for relative and absolute quantitation) analysis for normal and autistic children's plasma of the same age group. RESULTS The results show that 24 differentially expressed proteins were identified between autistic subjects and controls. For the first time, differential expression of complement C5 (C5) and fermitin family homolog 3 (FERMT3) are related to autism. Five proteins, that is, complement C3 (C3), C5, integrin alpha-IIb (ITGA2B), talin-1 (TLN1), and vitamin D-binding protein (GC) were validated via enzyme-linked immunosorbent assay (ELISA). By ROC (receiver operating characteristic) analysis, combinations of these five proteins C3, C5, GC, ITGA2B, and TLN1 distinguished autistic children from healthy controls with a high AUC (area under the ROC curve) value (0.982, 95% CI, 0.957-1.000, p < 0.000). CONCLUSION These above described proteins are found involved in different pathways that have previously been linked to the pathophysiology of autism spectrum disorders (ASDs). The results strongly support that focal adhesions, acting cytoskeleton, cell adhesion, motility and migration, synaptogenesis, and complement system are involved in the pathogenesis of autism, and highlight the important role of platelet function in the pathophysiology of autism.
Collapse
Affiliation(s)
- Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China
| | - Kaoyuan Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China
| | - Chengyun Feng
- Maternal and Child Health Hospital of Baoan, Shenzhen, P. R. China
| | - Youjiao Chen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China.,Xiang Ya Changde Hospital, Changde City, Hunan Province, P. R. China
| | - Shuiming Li
- College of Life Science and Oceanography, Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen University, Shenzhen, P. R. China
| | - Javed Iqbal
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China
| | - Liping Liao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China
| | - Yuxi Zhao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China
| | - Jian Zhai
- Maternal and Child Health Hospital of Baoan, Shenzhen, P. R. China
| |
Collapse
|
25
|
Fasoracetam in adolescents with ADHD and glutamatergic gene network variants disrupting mGluR neurotransmitter signaling. Nat Commun 2018; 9:4. [PMID: 29339723 PMCID: PMC5770454 DOI: 10.1038/s41467-017-02244-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 11/15/2017] [Indexed: 11/08/2022] Open
Abstract
The glutamatergic neurotransmitter system may play an important role in attention-deficit hyperactivity disorder (ADHD). This 5-week, open-label, single-blind, placebo-controlled study reports the safety, pharmacokinetics and responsiveness of the metabotropic glutamate receptor (mGluR) activator fasoracetam (NFC-1), in 30 adolescents, age 12-17 years with ADHD, harboring mutations in mGluR network genes. Mutation status was double-blinded. A single-dose pharmacokinetic profiling from 50-800 mg was followed by a single-blind placebo at week 1 and subsequent symptom-driven dose advancement up to 400 mg BID for 4 weeks. NFC-1 treatment resulted in significant improvement. Mean Clinical Global Impressions-Improvement (CGI-I) and Severity (CGI-S) scores were, respectively, 3.79 at baseline vs. 2.33 at week 5 (P < 0.001) and 4.83 at baseline vs. 3.86 at week 5 (P < 0.001). Parental Vanderbilt scores showed significant improvement for subjects with mGluR Tier 1 variants (P < 0.035). There were no differences in the incidence of adverse events between placebo week and weeks on active drug. The trial is registered at https://clinicaltrials.gov/ct2/show/study/NCT02286817 .
Collapse
|
26
|
Rappoport N, Toung J, Hadley D, Wong RJ, Fujioka K, Reuter J, Abbott CW, Oh S, Hu D, Eng C, Huntsman S, Bodian DL, Niederhuber JE, Hong X, Zhang G, Sikora-Wohfeld W, Gignoux CR, Wang H, Oehlert J, Jelliffe-Pawlowski LL, Gould JB, Darmstadt GL, Wang X, Bustamante CD, Snyder MP, Ziv E, Patsopoulos NA, Muglia LJ, Burchard E, Shaw GM, O'Brodovich HM, Stevenson DK, Butte AJ, Sirota M. A genome-wide association study identifies only two ancestry specific variants associated with spontaneous preterm birth. Sci Rep 2018; 8:226. [PMID: 29317701 PMCID: PMC5760643 DOI: 10.1038/s41598-017-18246-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/07/2017] [Indexed: 01/19/2023] Open
Abstract
Preterm birth (PTB), or the delivery prior to 37 weeks of gestation, is a significant cause of infant morbidity and mortality. Although twin studies estimate that maternal genetic contributions account for approximately 30% of the incidence of PTB, and other studies reported fetal gene polymorphism association, to date no consistent associations have been identified. In this study, we performed the largest reported genome-wide association study analysis on 1,349 cases of PTB and 12,595 ancestry-matched controls from the focusing on genomic fetal signals. We tested over 2 million single nucleotide polymorphisms (SNPs) for associations with PTB across five subpopulations: African (AFR), the Americas (AMR), European, South Asian, and East Asian. We identified only two intergenic loci associated with PTB at a genome-wide level of significance: rs17591250 (P = 4.55E-09) on chromosome 1 in the AFR population and rs1979081 (P = 3.72E-08) on chromosome 8 in the AMR group. We have queried several existing replication cohorts and found no support of these associations. We conclude that the fetal genetic contribution to PTB is unlikely due to single common genetic variant, but could be explained by interactions of multiple common variants, or of rare variants affected by environmental influences, all not detectable using a GWAS alone.
Collapse
Affiliation(s)
- Nadav Rappoport
- Institute for Computational Health Sciences, University of California, San Francisco, 94143, CA, USA.,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan Toung
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Dexter Hadley
- Institute for Computational Health Sciences, University of California, San Francisco, 94143, CA, USA.,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kazumichi Fujioka
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jason Reuter
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Charles W Abbott
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sam Oh
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Donglei Hu
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Celeste Eng
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Scott Huntsman
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Dale L Bodian
- Inova Translational Medicine Institute, Inova Health System, Falls Church, VA, USA
| | - John E Niederhuber
- Inova Translational Medicine Institute, Inova Health System, Falls Church, VA, USA.,Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Xiumei Hong
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ge Zhang
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | | | - Hui Wang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - John Oehlert
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jeffrey B Gould
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Gary L Darmstadt
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Carlos D Bustamante
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Elad Ziv
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Nikolaos A Patsopoulos
- Systems Biology and Computer Science Program, Ann Romney Center of Neurological Diseases, Department of Neurology, Division of Genetics, Brigham & Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Louis J Muglia
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Esteban Burchard
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Hugh M O'Brodovich
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Atul J Butte
- Institute for Computational Health Sciences, University of California, San Francisco, 94143, CA, USA. .,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA. .,Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA.
| | - Marina Sirota
- Institute for Computational Health Sciences, University of California, San Francisco, 94143, CA, USA. .,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA. .,Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
27
|
Zhang Y, Liu Y, Zarrei M, Tong W, Dong R, Wang Y, Zhang H, Yang X, MacDonald JR, Uddin M, Scherer SW, Gai Z. Association of IMMP2L deletions with autism spectrum disorder: A trio family study and meta-analysis. Am J Med Genet B Neuropsychiatr Genet 2018; 177:93-100. [PMID: 29152845 DOI: 10.1002/ajmg.b.32608] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 08/28/2017] [Accepted: 10/02/2017] [Indexed: 11/10/2022]
Abstract
IMMP2L, the gene encoding the inner mitochondrial membrane peptidase subunit 2-like protein, has been reported as a candidate gene for Tourette syndrome, autism spectrum disorder (ASD) and additional neurodevelopmental disorders. Here we genotyped 100 trio families with an index proband with autism spectrum disorder in Han Chinese population and found three cases with rare exonic IMMP2L deletions. We have conducted a comprehensive meta-analysis to quantify the association of IMMP2L deletions with ASD using 5,568 cases and 10,279 controls. While the IMMP2L deletions carried non-recurrent breakpoints, in contrast to previous reports, our meta-analysis found no evidence of association (P > 0.05) between IMMP2L deletions and ASD. We also observed common exonic deletions impacting IMMP2L in a separate control (5,971 samples) cohort where subjects were screened for psychiatric conditions. This is the first systematic review and meta-analysis regarding the effect of IMMP2L deletions on ASD, but further investigations in different populations, especially Chinese population may be still needed to confirm our results.
Collapse
Affiliation(s)
- Yanqing Zhang
- Pediatric Health Care Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Yi Liu
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Mehdi Zarrei
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Winnie Tong
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Rui Dong
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Ying Wang
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Haiyan Zhang
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Xiaomeng Yang
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Jeffrey R MacDonald
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Mohammed Uddin
- Mohammed Bin Rashid University of Medicine and Health Sciences, College of Medicine, Dubai, UAE
| | - Stephen W Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada.,McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Zhongtao Gai
- Pediatric Health Care Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China.,Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| |
Collapse
|
28
|
Glessner JT, Li J, Wang D, March M, Lima L, Desai A, Hadley D, Kao C, Gur RE, Cohen N, Sleiman PMA, Li Q, Hakonarson H. Copy number variation meta-analysis reveals a novel duplication at 9p24 associated with multiple neurodevelopmental disorders. Genome Med 2017; 9:106. [PMID: 29191242 PMCID: PMC5709845 DOI: 10.1186/s13073-017-0494-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/14/2017] [Indexed: 12/24/2022] Open
Abstract
Background Neurodevelopmental and neuropsychiatric disorders represent a wide spectrum of heterogeneous yet inter-related disease conditions. The overlapping clinical presentations of these diseases suggest a shared genetic etiology. We aim to identify shared structural variants spanning the spectrum of five neuropsychiatric disorders. Methods We investigated copy number variations (CNVs) in five cohorts, including schizophrenia (SCZ), bipolar disease (BD), autism spectrum disorders (ASD), attention deficit hyperactivity disorder (ADHD), and depression, from 7849 cases and 10,799 controls. CNVs were called based on intensity data from genome-wide SNP arrays and CNV frequency was compared between cases and controls in each disease cohort separately. Meta-analysis was performed via a gene-based approach. Quantitative PCR (qPCR) was employed to validate novel significant loci. Results In our meta-analysis, two genes containing CNVs with exonic overlap reached genome-wide significance threshold of meta P value < 9.4 × 10−6 for deletions and 7.5 × 10−6 for duplications. We observed significant overlap between risk CNV loci across cohorts. In addition, we identified novel significant associations of DOCK8/KANK1 duplications (meta P value = 7.5 × 10−7) across all cohorts, and further validated the CNV region with qPCR. Conclusions In the first large scale meta-analysis of CNVs across multiple neurodevelopmental/psychiatric diseases, we uncovered novel significant associations of structural variants in the locus of DOCK8/KANK1 shared by five diseases, suggesting common etiology of these clinically distinct neurodevelopmental conditions. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0494-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joseph T Glessner
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Jin Li
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Cell Biology, Tianjin Medical University, Tianjin, China
| | - Dai Wang
- Janssen Research & Development, LLC, Raritan, NJ, 08869, USA
| | - Michael March
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Leandro Lima
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Akshatha Desai
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Dexter Hadley
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Charlly Kao
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Raquel E Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nadine Cohen
- Janssen Research & Development, LLC, Raritan, NJ, 08869, USA
| | - Patrick M A Sleiman
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Qingqin Li
- Janssen Research & Development, LLC, Titusville, NJ, 08560, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA. .,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. .,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
| | | |
Collapse
|
29
|
Clements CC, Wenger TL, Zoltowski AR, Bertollo JR, Miller JS, de Marchena AB, Mitteer LM, Carey JC, Yerys BE, Zackai EH, Emanuel BS, McDonald-McGinn DM, Schultz RT. Critical region within 22q11.2 linked to higher rate of autism spectrum disorder. Mol Autism 2017; 8:58. [PMID: 29090080 PMCID: PMC5658953 DOI: 10.1186/s13229-017-0171-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/27/2017] [Indexed: 11/26/2022] Open
Abstract
Background Previous studies have reported no clear critical region for medical comorbidities in children with deletions or duplications of 22q11.2. The purpose of this study was to evaluate whether individuals with small nested deletions or duplications of the LCR-A to B region of 22q11.2 show an elevated rate of autism spectrum disorder (ASD) compared to individuals with deletions or duplications that do not include this region. Methods We recruited 46 patients with nested deletions (n = 33) or duplications (n = 13) of 22q11.2, including LCR-A to B (ndel = 11), LCR-A to C (ndel = 4), LCR-B to D (ndel = 14; ndup = 8), LCR-C to D (ndel = 4; ndup = 2), and smaller nested regions (n = 3). Parent questionnaire, record review, and, for a subset, in-person evaluation were used for ASD diagnostic classification. Rates of ASD in individuals with involvement of LCR-B to LCR-D were compared with Fisher’s exact test to LCR-A to LCR-B for deletions, and to a previously published sample of LCR-A to LCR-D for duplications. The rates of medical comorbidities and psychiatric diagnoses were determined from questionnaires and chart review. We also report group mean differences on psychiatric questionnaires. Results Individuals with deletions involving LCR-A to B showed a 39–44% rate of ASD compared to 0% in individuals whose deletions did not involve LCR-A to B. We observed similar rates of medical comorbidities in individuals with involvement of LCR-A to B and LCR-B to D for both duplications and deletions, consistent with prior studies. Conclusions Children with nested deletions of 22q11.2 may be at greater risk for autism spectrum disorder if the region includes LCR-A to LCR-B. Replication is needed. Electronic supplementary material The online version of this article (10.1186/s13229-017-0171-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caitlin C Clements
- Center for Autism Research, The Children's Hospital of Philadelphia, 2716 South Street, Philadelphia, PA 19104 USA.,Department of Psychology, University of Pennsylvania, 3720 Walnut Street, Philadelphia, PA 19104 USA
| | - Tara L Wenger
- Center for Autism Research, The Children's Hospital of Philadelphia, 2716 South Street, Philadelphia, PA 19104 USA.,Department of Pediatrics, Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA 98105 USA
| | - Alisa R Zoltowski
- Center for Autism Research, The Children's Hospital of Philadelphia, 2716 South Street, Philadelphia, PA 19104 USA.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, 465 21st Avenue South, Nashville, TN 37232 USA
| | - Jennifer R Bertollo
- Center for Autism Research, The Children's Hospital of Philadelphia, 2716 South Street, Philadelphia, PA 19104 USA
| | - Judith S Miller
- Center for Autism Research, The Children's Hospital of Philadelphia, 2716 South Street, Philadelphia, PA 19104 USA.,Department of Psychiatry, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104 USA
| | - Ashley B de Marchena
- Center for Autism Research, The Children's Hospital of Philadelphia, 2716 South Street, Philadelphia, PA 19104 USA.,Department of Behavioral and Social Science, University of the Sciences, 600 South 43rd Street, Philadelphia, PA 19104 USA
| | - Lauren M Mitteer
- Department of Pediatrics, The Children's Hospital of Philadelphia, 3400 Civic Center Boulevard, Philadelphia, PA 19104 USA
| | - John C Carey
- Department of Pediatrics, University of Utah, Salt Lake City, UT 84108 USA
| | - Benjamin E Yerys
- Center for Autism Research, The Children's Hospital of Philadelphia, 2716 South Street, Philadelphia, PA 19104 USA.,Department of Psychiatry, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104 USA
| | - Elaine H Zackai
- Department of Pediatrics, The Children's Hospital of Philadelphia, 3400 Civic Center Boulevard, Philadelphia, PA 19104 USA
| | - Beverly S Emanuel
- Department of Pediatrics, The Children's Hospital of Philadelphia, 3400 Civic Center Boulevard, Philadelphia, PA 19104 USA
| | - Donna M McDonald-McGinn
- Department of Pediatrics, The Children's Hospital of Philadelphia, 3400 Civic Center Boulevard, Philadelphia, PA 19104 USA
| | - Robert T Schultz
- Center for Autism Research, The Children's Hospital of Philadelphia, 2716 South Street, Philadelphia, PA 19104 USA.,Department of Psychiatry, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104 USA.,Department of Pediatrics, The Children's Hospital of Philadelphia, 3400 Civic Center Boulevard, Philadelphia, PA 19104 USA
| |
Collapse
|
30
|
Hadley D, Pan J, El-Sayed O, Aljabban J, Aljabban I, Azad TD, Hadied MO, Raza S, Rayikanti BA, Chen B, Paik H, Aran D, Spatz J, Himmelstein D, Panahiazar M, Bhattacharya S, Sirota M, Musen MA, Butte AJ. Precision annotation of digital samples in NCBI's gene expression omnibus. Sci Data 2017; 4:170125. [PMID: 28925997 PMCID: PMC5604135 DOI: 10.1038/sdata.2017.125] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 07/28/2017] [Indexed: 12/16/2022] Open
Abstract
The Gene Expression Omnibus (GEO) contains more than two million digital samples from functional genomics experiments amassed over almost two decades. However, individual sample meta-data remains poorly described by unstructured free text attributes preventing its largescale reanalysis. We introduce the Search Tag Analyze Resource for GEO as a web application (http://STARGEO.org) to curate better annotations of sample phenotypes uniformly across different studies, and to use these sample annotations to define robust genomic signatures of disease pathology by meta-analysis. In this paper, we target a small group of biomedical graduate students to show rapid crowd-curation of precise sample annotations across all phenotypes, and we demonstrate the biological validity of these crowd-curated annotations for breast cancer. STARGEO.org makes GEO data findable, accessible, interoperable and reusable (i.e., FAIR) to ultimately facilitate knowledge discovery. Our work demonstrates the utility of crowd-curation and interpretation of open ‘big data’ under FAIR principles as a first step towards realizing an ideal paradigm of precision medicine.
Collapse
Affiliation(s)
- Dexter Hadley
- Institute for Computational Health Sciences, University of California, San Francisco, California 94158, USA
| | - James Pan
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Osama El-Sayed
- University of Illinois College of Medicine, Chicago, Illinois 60612, USA
| | - Jihad Aljabban
- Harvard Medical School Department of Immunology, Harvard University, Boston, Massachusetts 02115, USA
| | - Imad Aljabban
- Harvard Medical School Department of Immunology, Harvard University, Boston, Massachusetts 02115, USA
| | - Tej D Azad
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Mohamad O Hadied
- Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | - Shuaib Raza
- Yale School of Medicine, Yale University, New Haven, Connecticut 06519, USA
| | | | - Bin Chen
- Institute for Computational Health Sciences, University of California, San Francisco, California 94158, USA
| | - Hyojung Paik
- Institute for Computational Health Sciences, University of California, San Francisco, California 94158, USA
| | - Dvir Aran
- Institute for Computational Health Sciences, University of California, San Francisco, California 94158, USA
| | - Jordan Spatz
- Institute for Computational Health Sciences, University of California, San Francisco, California 94158, USA
| | - Daniel Himmelstein
- Program in Biological &Medical Informatics, University of California, San Francisco, CA 94158, USA
| | - Maryam Panahiazar
- Institute for Computational Health Sciences, University of California, San Francisco, California 94158, USA
| | - Sanchita Bhattacharya
- Institute for Computational Health Sciences, University of California, San Francisco, California 94158, USA
| | - Marina Sirota
- Institute for Computational Health Sciences, University of California, San Francisco, California 94158, USA
| | - Mark A Musen
- Stanford Center for Biomedical Informatics Research, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Atul J Butte
- Institute for Computational Health Sciences, University of California, San Francisco, California 94158, USA
| |
Collapse
|
31
|
Sharp BM. Basolateral amygdala and stress-induced hyperexcitability affect motivated behaviors and addiction. Transl Psychiatry 2017; 7:e1194. [PMID: 28786979 PMCID: PMC5611728 DOI: 10.1038/tp.2017.161] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/16/2017] [Accepted: 06/08/2017] [Indexed: 12/11/2022] Open
Abstract
The amygdala integrates and processes incoming information pertinent to reward and to emotions such as fear and anxiety that promote survival by warning of potential danger. Basolateral amygdala (BLA) communicates bi-directionally with brain regions affecting cognition, motivation and stress responses including prefrontal cortex, hippocampus, nucleus accumbens and hindbrain regions that trigger norepinephrine-mediated stress responses. Disruption of intrinsic amygdala and BLA regulatory neurocircuits is often caused by dysfunctional neuroplasticity frequently due to molecular alterations in local GABAergic circuits and principal glutamatergic output neurons. Changes in local regulation of BLA excitability underlie behavioral disturbances characteristic of disorders including post-traumatic stress syndrome (PTSD), autism, attention-deficit hyperactivity disorder (ADHD) and stress-induced relapse to drug use. In this Review, we discuss molecular mechanisms and neural circuits that regulate physiological and stress-induced dysfunction of BLA/amygdala and its principal output neurons. We consider effects of stress on motivated behaviors that depend on BLA; these include drug taking and drug seeking, with emphasis on nicotine-dependent behaviors. Throughout, we take a translational approach by integrating decades of addiction research on animal models and human trials. We show that changes in BLA function identified in animal addiction models illuminate human brain imaging and behavioral studies by more precisely delineating BLA mechanisms. In summary, BLA is required to promote responding for natural reward and respond to second-order drug-conditioned cues; reinstate cue-dependent drug seeking; express stress-enhanced reacquisition of nicotine intake; and drive anxiety and fear. Converging evidence indicates that chronic stress causes BLA principal output neurons to become hyperexcitable.
Collapse
Affiliation(s)
- B M Sharp
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
32
|
Whole exome sequencing reveals inherited and de novo variants in autism spectrum disorder: a trio study from Saudi families. Sci Rep 2017; 7:5679. [PMID: 28720891 PMCID: PMC5515956 DOI: 10.1038/s41598-017-06033-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 06/06/2017] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with genetic and clinical heterogeneity. The interplay of de novo and inherited rare variants has been suspected in the development of ASD. Here, we applied whole exome sequencing (WES) on 19 trios from singleton Saudi families with ASD. We developed an analysis pipeline that allows capturing both de novo and inherited rare variants predicted to be deleterious. A total of 47 unique rare variants were detected in 17 trios including 38 which are newly discovered. The majority were either autosomal recessive or X-linked. Our pipeline uncovered variants in 15 ASD-candidate genes, including 5 (GLT8D1, HTATSF1, OR6C65, ITIH6 and DDX26B) that have not been reported in any human condition. The remaining variants occurred in genes formerly associated with ASD or other neurological disorders. Examples include SUMF1, KDM5B and MXRA5 (Known-ASD genes), PRODH2 and KCTD21 (implicated in schizophrenia), as well as USP9X and SMS (implicated in intellectual disability). Consistent with expectation and previous studies, most of the genes implicated herein are enriched for biological processes pertaining to neuronal function. Our findings underscore the private and heterogeneous nature of the genetic architecture of ASD even in a population with high consanguinity rates.
Collapse
|
33
|
Abstract
Human genetic studies have been the driving force in bringing to light the underlying biology of psychiatric conditions. As these studies fill in the gaps in our knowledge of the mechanisms at play, we will be better equipped to design therapies in rational and targeted ways, or repurpose existing therapies in previously unanticipated ways. This review is intended for those unfamiliar with psychiatric genetics as a field and provides a primer on different modes of genetic variation, the technologies currently used to probe them, and concepts that provide context for interpreting the gene-phenotype relationship. Like other subfields in human genetics, psychiatric genetics is moving from microarray technology to sequencing-based approaches as barriers of cost and expertise are removed, and the ramifications of this transition are discussed here. A summary is then given of recent genetic discoveries in a number of neuropsychiatric conditions, with particular emphasis on neurodevelopmental conditions. The general impact of genetics on drug development has been to underscore the extensive etiological heterogeneity in seemingly cohesive diagnostic categories. Consequently, the path forward is not in therapies hoping to reach large swaths of patients sharing a clinically defined diagnosis, but rather in targeting patients belonging to specific "biotypes" defined through a combination of objective, quantifiable data, including genotype.
Collapse
Affiliation(s)
- Jacob J Michaelson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
- Department of Biomedical Engineering, University of Iowa College of Engineering, Iowa City, IA, USA.
- Department of Communication Sciences and Disorders, University of Iowa College of Liberal Arts and Sciences, Iowa City, IA, USA.
- Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA, USA.
- Genetics Cluster Initiative, University of Iowa, Iowa City, IA, USA.
- The DeLTA Center, University of Iowa, Iowa City, IA, USA.
- University of Iowa Informatics Initiative, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
34
|
Tetrahelical structural family adopted by AGCGA-rich regulatory DNA regions. Nat Commun 2017; 8:15355. [PMID: 28513602 PMCID: PMC5442326 DOI: 10.1038/ncomms15355] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/23/2017] [Indexed: 12/13/2022] Open
Abstract
Here we describe AGCGA-quadruplexes, an unexpected addition to the well-known tetrahelical families, G-quadruplexes and i-motifs, that have been a focus of intense research due to their potential biological impact in G- and C-rich DNA regions, respectively. High-resolution structures determined by solution-state nuclear magnetic resonance (NMR) spectroscopy demonstrate that AGCGA-quadruplexes comprise four 5′-AGCGA-3′ tracts and are stabilized by G-A and G-C base pairs forming GAGA- and GCGC-quartets, respectively. Residues in the core of the structure are connected with edge-type loops. Sequences of alternating 5′-AGCGA-3′ and 5′-GGG-3′ repeats could be expected to form G-quadruplexes, but are shown herein to form AGCGA-quadruplexes instead. Unique structural features of AGCGA-quadruplexes together with lower sensitivity to cation and pH variation imply their potential biological relevance in regulatory regions of genes responsible for basic cellular processes that are related to neurological disorders, cancer and abnormalities in bone and cartilage development. DNA tetrahelical structures such as G-quadruplexes are known to play important roles in DNA replication and repair. Here the authors present the structure of 5′-AGCGA-3′-quadruplexes enriched in genetic regulatory regions.
Collapse
|
35
|
Delineating the Common Biological Pathways Perturbed by ASD's Genetic Etiology: Lessons from Network-Based Studies. Int J Mol Sci 2017; 18:ijms18040828. [PMID: 28420080 PMCID: PMC5412412 DOI: 10.3390/ijms18040828] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 12/26/2022] Open
Abstract
In recent decades it has become clear that Autism Spectrum Disorder (ASD) possesses a diverse and heterogeneous genetic etiology. Aberrations in hundreds of genes have been associated with ASD so far, which include both rare and common variations. While one may expect that these genes converge on specific common molecular pathways, which drive the development of the core ASD characteristics, the task of elucidating these common molecular pathways has been proven to be challenging. Several studies have combined genetic analysis with bioinformatical techniques to uncover molecular mechanisms that are specifically targeted by autism-associated genetic aberrations. Recently, several analysis have suggested that particular signaling mechanisms, including the Wnt and Ca2+/Calmodulin-signaling pathways are often targeted by autism-associated mutations. In this review, we discuss several studies that determine specific molecular pathways affected by autism-associated mutations, and then discuss more in-depth into the biological roles of a few of these pathways, and how they may be involved in the development of ASD. Considering that these pathways may be targeted by specific pharmacological intervention, they may prove to be important therapeutic targets for the treatment of ASD.
Collapse
|
36
|
Stergiakouli E, Davey Smith G, Martin J, Skuse DH, Viechtbauer W, Ring SM, Ronald A, Evans DE, Fisher SE, Thapar A, St Pourcain B. Shared genetic influences between dimensional ASD and ADHD symptoms during child and adolescent development. Mol Autism 2017; 8:18. [PMID: 28392908 PMCID: PMC5379648 DOI: 10.1186/s13229-017-0131-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 03/14/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Shared genetic influences between attention-deficit/hyperactivity disorder (ADHD) symptoms and autism spectrum disorder (ASD) symptoms have been reported. Cross-trait genetic relationships are, however, subject to dynamic changes during development. We investigated the continuity of genetic overlap between ASD and ADHD symptoms in a general population sample during childhood and adolescence. We also studied uni- and cross-dimensional trait-disorder links with respect to genetic ADHD and ASD risk. METHODS Social-communication difficulties (N ≤ 5551, Social and Communication Disorders Checklist, SCDC) and combined hyperactive-impulsive/inattentive ADHD symptoms (N ≤ 5678, Strengths and Difficulties Questionnaire, SDQ-ADHD) were repeatedly measured in a UK birth cohort (ALSPAC, age 7 to 17 years). Genome-wide summary statistics on clinical ASD (5305 cases; 5305 pseudo-controls) and ADHD (4163 cases; 12,040 controls/pseudo-controls) were available from the Psychiatric Genomics Consortium. Genetic trait variances and genetic overlap between phenotypes were estimated using genome-wide data. RESULTS In the general population, genetic influences for SCDC and SDQ-ADHD scores were shared throughout development. Genetic correlations across traits reached a similar strength and magnitude (cross-trait rg ≤ 1, pmin = 3 × 10-4) as those between repeated measures of the same trait (within-trait rg ≤ 0.94, pmin = 7 × 10-4). Shared genetic influences between traits, especially during later adolescence, may implicate variants in K-RAS signalling upregulated genes (p-meta = 6.4 × 10-4). Uni-dimensionally, each population-based trait mapped to the expected behavioural continuum: risk-increasing alleles for clinical ADHD were persistently associated with SDQ-ADHD scores throughout development (marginal regression R2 = 0.084%). An age-specific genetic overlap between clinical ASD and social-communication difficulties during childhood was also shown, as per previous reports. Cross-dimensionally, however, neither SCDC nor SDQ-ADHD scores were linked to genetic risk for disorder. CONCLUSIONS In the general population, genetic aetiologies between social-communication difficulties and ADHD symptoms are shared throughout child and adolescent development and may implicate similar biological pathways that co-vary during development. Within both the ASD and the ADHD dimension, population-based traits are also linked to clinical disorder, although much larger clinical discovery samples are required to reliably detect cross-dimensional trait-disorder relationships.
Collapse
Affiliation(s)
- Evie Stergiakouli
- MRC Integrative Epidemiology Unit (MRC IEU), University of Bristol, Bristol, UK
- School of Oral and Dental Sciences, University of Bristol, Bristol, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (MRC IEU), University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Joanna Martin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - David H. Skuse
- Institute of Child Health, University College London, London, UK
| | - Wolfgang Viechtbauer
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
| | - Susan M. Ring
- MRC Integrative Epidemiology Unit (MRC IEU), University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Angelica Ronald
- Department of Psychological Sciences, Birkbeck, University of London, London, UK
| | - David E. Evans
- MRC Integrative Epidemiology Unit (MRC IEU), University of Bristol, Bristol, UK
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Australia
| | - Simon E. Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Anita Thapar
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Beate St Pourcain
- MRC Integrative Epidemiology Unit (MRC IEU), University of Bristol, Bristol, UK
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| |
Collapse
|
37
|
Genome-wide copy number variation analysis in a Chinese autism spectrum disorder cohort. Sci Rep 2017; 7:44155. [PMID: 28281572 PMCID: PMC5345089 DOI: 10.1038/srep44155] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/03/2017] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder (ASD) describes a group of neurodevelopmental disorders with high heritability, although the underlying genetic determinants of ASDs remain largely unknown. Large-scale whole-genome studies of copy number variation in Han Chinese samples are still lacking. We performed a genome-wide copy number variation analysis of 343 ASD trios, 203 patients with sporadic cases and 988 controls in a Chinese population using Illumina genotyping platforms to identify CNVs and related genes that may contribute to ASD risk. We identified 32 rare CNVs larger than 1 Mb in 31 patients. ASD patients were found to carry a higher global burden of rare, large CNVs than controls. Recurrent de novo or case-private CNVs were found at 15q11-13, Xp22.3, 15q13.1–13.2, 3p26.3 and 2p12. The de novo 15q11–13 duplication was more prevalent in this Chinese population than in those with European ancestry. Several genes, including GRAMD2 and STAM, were implicated as novel ASD risk genes when integrating whole-genome CNVs and whole-exome sequencing data. We also identified several CNVs that include known ASD genes (SHANK3, CDH10, CSMD1) or genes involved in nervous system development (NYAP2, ST6GAL2, GRM6). Besides, our study also implicated Contactins-NYAPs-WAVE1 pathway in ASD pathogenesis. Our findings identify ASD-related CNVs in a Chinese population and implicate novel ASD risk genes and related pathway for further study.
Collapse
|
38
|
C Yuen RK, Merico D, Bookman M, L Howe J, Thiruvahindrapuram B, Patel RV, Whitney J, Deflaux N, Bingham J, Wang Z, Pellecchia G, Buchanan JA, Walker S, Marshall CR, Uddin M, Zarrei M, Deneault E, D'Abate L, Chan AJS, Koyanagi S, Paton T, Pereira SL, Hoang N, Engchuan W, Higginbotham EJ, Ho K, Lamoureux S, Li W, MacDonald JR, Nalpathamkalam T, Sung WWL, Tsoi FJ, Wei J, Xu L, Tasse AM, Kirby E, Van Etten W, Twigger S, Roberts W, Drmic I, Jilderda S, Modi BM, Kellam B, Szego M, Cytrynbaum C, Weksberg R, Zwaigenbaum L, Woodbury-Smith M, Brian J, Senman L, Iaboni A, Doyle-Thomas K, Thompson A, Chrysler C, Leef J, Savion-Lemieux T, Smith IM, Liu X, Nicolson R, Seifer V, Fedele A, Cook EH, Dager S, Estes A, Gallagher L, Malow BA, Parr JR, Spence SJ, Vorstman J, Frey BJ, Robinson JT, Strug LJ, Fernandez BA, Elsabbagh M, Carter MT, Hallmayer J, Knoppers BM, Anagnostou E, Szatmari P, Ring RH, Glazer D, Pletcher MT, Scherer SW. Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder. Nat Neurosci 2017; 20:602-611. [PMID: 28263302 DOI: 10.1038/nn.4524] [Citation(s) in RCA: 554] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/01/2017] [Indexed: 12/13/2022]
Abstract
We are performing whole-genome sequencing of families with autism spectrum disorder (ASD) to build a resource (MSSNG) for subcategorizing the phenotypes and underlying genetic factors involved. Here we report sequencing of 5,205 samples from families with ASD, accompanied by clinical information, creating a database accessible on a cloud platform and through a controlled-access internet portal. We found an average of 73.8 de novo single nucleotide variants and 12.6 de novo insertions and deletions or copy number variations per ASD subject. We identified 18 new candidate ASD-risk genes and found that participants bearing mutations in susceptibility genes had significantly lower adaptive ability (P = 6 × 10-4). In 294 of 2,620 (11.2%) of ASD cases, a molecular basis could be determined and 7.2% of these carried copy number variations and/or chromosomal abnormalities, emphasizing the importance of detecting all forms of genetic variation as diagnostic and therapeutic targets in ASD.
Collapse
Affiliation(s)
- Ryan K C Yuen
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Daniele Merico
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Deep Genomics Inc., Toronto, Canada
| | - Matt Bookman
- Google, Mountain View, California, USA.,Verily Life Sciences, South San Francisco, California, USA
| | - Jennifer L Howe
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Bhooma Thiruvahindrapuram
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Rohan V Patel
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Joe Whitney
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Nicole Deflaux
- Google, Mountain View, California, USA.,Verily Life Sciences, South San Francisco, California, USA
| | - Jonathan Bingham
- Google, Mountain View, California, USA.,Verily Life Sciences, South San Francisco, California, USA
| | - Zhuozhi Wang
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Giovanna Pellecchia
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Janet A Buchanan
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Susan Walker
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Christian R Marshall
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
| | - Mohammed Uddin
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Mehdi Zarrei
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Eric Deneault
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Lia D'Abate
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Ada J S Chan
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Stephanie Koyanagi
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Tara Paton
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Sergio L Pereira
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Ny Hoang
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
| | - Worrawat Engchuan
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Edward J Higginbotham
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Karen Ho
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Sylvia Lamoureux
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Weili Li
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Jeffrey R MacDonald
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Thomas Nalpathamkalam
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Wilson W L Sung
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Fiona J Tsoi
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - John Wei
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Lizhen Xu
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Anne-Marie Tasse
- Public Population Project in Genomics and Society, McGill University, Montreal, Canada
| | - Emily Kirby
- Public Population Project in Genomics and Society, McGill University, Montreal, Canada
| | | | | | - Wendy Roberts
- Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
| | - Irene Drmic
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
| | - Sanne Jilderda
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
| | - Bonnie MacKinnon Modi
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
| | - Barbara Kellam
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Michael Szego
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Dalla Lana School of Public Health and the Department of Family and Community Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Cheryl Cytrynbaum
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Dalla Lana School of Public Health and the Department of Family and Community Medicine, University of Toronto, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Canada
| | - Rosanna Weksberg
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Canada
| | | | - Marc Woodbury-Smith
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| | - Jessica Brian
- Bloorview Research Institute, University of Toronto, Toronto, Canada.
| | - Lili Senman
- Bloorview Research Institute, University of Toronto, Toronto, Canada.
| | - Alana Iaboni
- Bloorview Research Institute, University of Toronto, Toronto, Canada.
| | | | - Ann Thompson
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| | - Christina Chrysler
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| | - Jonathan Leef
- Bloorview Research Institute, University of Toronto, Toronto, Canada.
| | | | - Isabel M Smith
- Departments of Pediatrics and of Psychology &Neuroscience, Dalhousie University and Autism Research Centre, IWK Health Centre, Halifax, Canada
| | - Xudong Liu
- Department of Psychiatry, Queen's University, Kinston, Canada
| | - Rob Nicolson
- Children's Health Research Institute, London, Ontario, Canada.,Western University, London, Ontario, Canada
| | | | | | - Edwin H Cook
- Institute for Juvenile Research, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Stephen Dager
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Annette Estes
- Department of Speech and Hearing Sciences, University of Washington, Seattle, Washington, USA
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Beth A Malow
- Sleep Disorders Division, Department of Neurology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jeremy R Parr
- Institute of Neuroscience, Newcastle University, Newcastle Upon Tyne, UK
| | - Sarah J Spence
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jacob Vorstman
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Brendan J Frey
- Deep Genomics Inc., Toronto, Canada.,Department of Electrical and Computer Engineering and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - James T Robinson
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Lisa J Strug
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Canada
| | - Bridget A Fernandez
- Disciplines of Genetics and Medicine, Memorial University of Newfoundland and Provincial Medical Genetic Program, Eastern Health, St. John's, Canada
| | | | - Melissa T Carter
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Canada.,Regional Genetics Program, The Children's Hospital of Eastern Ontario, Ottawa, Canada
| | - Joachim Hallmayer
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, USA
| | | | | | - Peter Szatmari
- Child Youth and Family Services, Centre for Addiction and Mental Health, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada.,Department of Psychiatry, The Hospital for Sick Children, Toronto, Canada
| | - Robert H Ring
- Department of Pharmacology &Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - David Glazer
- Google, Mountain View, California, USA.,Verily Life Sciences, South San Francisco, California, USA
| | | | - Stephen W Scherer
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada.,McLaughlin Centre, University of Toronto, Toronto, Canada
| |
Collapse
|
39
|
Noroozi R, Taheri M, Movafagh A, Mirfakhraie R, Solgi G, Sayad A, Mazdeh M, Darvish H. Glutamate receptor, metabotropic 7 (GRM7) gene variations and susceptibility to autism: A case-control study. Autism Res 2016; 9:1161-1168. [DOI: 10.1002/aur.1640] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 04/04/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Rezvan Noroozi
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Mohammad Taheri
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Abolfazl Movafagh
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Reza Mirfakhraie
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine; Hamadan University of Medical Sciences; Hamadan Iran
| | - Arezou Sayad
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Mehrdokht Mazdeh
- Department of Neurology, School of Medicine; Hamadan University of Medical Sciences; Hamadan Iran
| | - Hossein Darvish
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
40
|
Antshel KM, Zhang-James Y, Wagner KE, Ledesma A, Faraone SV. An update on the comorbidity of ADHD and ASD: a focus on clinical management. Expert Rev Neurother 2016; 16:279-93. [PMID: 26807870 DOI: 10.1586/14737175.2016.1146591] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Attention deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) commonly co-occur. With the DSM-5, clinicians are permitted to make an ASD diagnosis in the context of ADHD. In earlier versions of the DSM, this was not acceptable. Both ASD and ADHD are reported to have had substantial increases in prevalence within the past 10 years. As a function of both the increased prevalence of both disorders as well as the ability to make an ASD diagnosis in ADHD, there has been a significant amount of research focusing on the comorbidity between ADHD and ASD in the past few years. Here, we provide an update on the biological, cognitive and behavioral overlap/distinctiveness between the two neurodevelopmental disorders with a focus on data published in the last four years. Treatment strategies for the comorbid condition as well as future areas of research and clinical need are discussed.
Collapse
Affiliation(s)
- Kevin M Antshel
- a Department of Psychology , Syracuse University , Syracuse , NY , USA.,b Department of Psychiatry & Behavioral Sciences , SUNY-Upstate Medical University , Syracuse , NY , USA
| | - Yanli Zhang-James
- b Department of Psychiatry & Behavioral Sciences , SUNY-Upstate Medical University , Syracuse , NY , USA
| | - Kayla E Wagner
- a Department of Psychology , Syracuse University , Syracuse , NY , USA
| | - Ana Ledesma
- a Department of Psychology , Syracuse University , Syracuse , NY , USA
| | - Stephen V Faraone
- b Department of Psychiatry & Behavioral Sciences , SUNY-Upstate Medical University , Syracuse , NY , USA.,c K.G. Jebsen Centre for Research on Neuropsychiatric Disorders , University of Bergen , Bergen , Norway.,d Department of Neuroscience and Physiology , SUNY-Upstate Medical University , Syracuse , NY , USA
| |
Collapse
|
41
|
Full-field electroretinogram in autism spectrum disorder. Doc Ophthalmol 2016; 132:83-99. [DOI: 10.1007/s10633-016-9529-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 02/02/2016] [Indexed: 11/25/2022]
|
42
|
Wenger TL, Kao C, McDonald-McGinn DM, Zackai EH, Bailey A, Schultz RT, Morrow BE, Emanuel BS, Hakonarson H. The Role of mGluR Copy Number Variation in Genetic and Environmental Forms of Syndromic Autism Spectrum Disorder. Sci Rep 2016; 6:19372. [PMID: 26781481 PMCID: PMC4726047 DOI: 10.1038/srep19372] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 11/18/2015] [Indexed: 11/09/2022] Open
Abstract
While abnormal signaling mediated through metabotropic glutamate receptor 5 (mGluR5) is involved in the pathophysiology of Autism Spectrum Disorder (ASD), Fragile X Syndrome and Tuberous Sclerosis, the role of other mGluRs and their associated signaling network genes in syndromic ASD is unknown. This study sought to determine whether mGluR Copy Number Variants (CNV's) were overrepresented in children with syndromic ASD and if mGluR "second hit" confers additional risk for ASD in 22q11.2 Deletion Syndrome (22q11DS). To determine whether mGluR network CNV'S are enriched in syndromic ASD, we examined microarrays from children with ASD (n = 539). Patient categorization (syndromic vs nonsyndromic) was done via blinded medical chart review in mGluR positive and randomly selected mGluR negative cases. 11.5% of ASD had mGluR CNV's vs. 3.2% in controls (p < 0.001). Syndromic ASD was more prevalent in children with mGluR CNVs (74% vs 16%, p < 0.001). A comparison cohort with 22q11DS (n = 25 with ASD, n = 50 without ASD), all haploinsufficient for mGluR network gene RANBP1, were evaluated for "second mGluR hits". 20% with 22q11.2DS + ASD had "second hits" in mGluR network genes vs 2% in 22q11.2DS-ASD (p < 0.014). We propose that altered RANBP1 expression may provide a mechanistic link for several seemingly unrelated genetic and environmental forms of ASD.
Collapse
Affiliation(s)
- Tara L. Wenger
- Seattle Children’s Hospital, Department of Pediatrics, Seattle, WA 98105 USA
- Children’s Hospital of Philadelphia, Department of Pediatrics, Philadelphia, PA 19104 USA
| | - Charlly Kao
- Children’s Hospital of Philadelphia, Department of Pediatrics, Philadelphia, PA 19104 USA
| | | | - Elaine H. Zackai
- Children’s Hospital of Philadelphia, Department of Pediatrics, Philadelphia, PA 19104 USA
| | - Alice Bailey
- Children’s Hospital of Philadelphia, Department of Pediatrics, Philadelphia, PA 19104 USA
| | - Robert T. Schultz
- Children’s Hospital of Philadelphia, Department of Pediatrics, Philadelphia, PA 19104 USA
| | - Bernice E. Morrow
- Albert Einstein College of Medicine, Department of Genetics, Bronx, NY 10461 USA
| | - Beverly S. Emanuel
- Children’s Hospital of Philadelphia, Department of Pediatrics, Philadelphia, PA 19104 USA
| | - Hakon Hakonarson
- Children’s Hospital of Philadelphia, Department of Pediatrics, Philadelphia, PA 19104 USA
| |
Collapse
|
43
|
Maruani A, Huguet G, Beggiato A, ElMaleh M, Toro R, Leblond CS, Mathieu A, Amsellem F, Lemière N, Verloes A, Leboyer M, Gillberg C, Bourgeron T, Delorme R. 11q24.2-25 micro-rearrangements in autism spectrum disorders: Relation to brain structures. Am J Med Genet A 2015; 167A:3019-30. [DOI: 10.1002/ajmg.a.37345] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 08/12/2015] [Indexed: 01/24/2023]
Affiliation(s)
- Anna Maruani
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Department of Child Psychiatry; Robert Debré Hospital, APHP; Paris France
| | - Guillaume Huguet
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Anita Beggiato
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Department of Child Psychiatry; Robert Debré Hospital, APHP; Paris France
| | - Monique ElMaleh
- Department of Radiology; Robert Debré Hospital, APHP; Paris France
| | - Roberto Toro
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Claire S. Leblond
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Alexandre Mathieu
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | | | - Nathalie Lemière
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Alain Verloes
- Department of Human Genetics; Robert Debré Hospital, APHP; Paris France
| | - Marion Leboyer
- INSERM U955, Team 15; Faculty of Medicine; Creteil France
- Department of Adult Psychiatry; Henri Mondor-Albert Chenevier Hospitals AP-HP; Creteil France
- Fondation FondaMental; French National Science Foundation; Creteil France
| | - Christopher Gillberg
- Gillberg Neuropsychiatry Centre; Gothenburg University; Göteborg Sweden
- Saint George's Hospital Medical School; London United Kingdom
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Diderot Paris 7; University Paris; Paris France
| | - Richard Delorme
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Department of Child Psychiatry; Robert Debré Hospital, APHP; Paris France
- Fondation FondaMental; French National Science Foundation; Creteil France
| |
Collapse
|
44
|
Niu W, Huang X, Yu T, Chen S, Li X, Wu X, Cao Y, Zhang R, Bi Y, Yang F, Wang L, Li W, Xu Y, He L, He G. Association study of GRM7 polymorphisms and schizophrenia in the Chinese Han population. Neurosci Lett 2015; 604:109-12. [PMID: 26254163 DOI: 10.1016/j.neulet.2015.07.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 11/26/2022]
|
45
|
Liu Y, Zhang Y, Zhao D, Dong R, Yang X, Tammimies K, Uddin M, Scherer SW, Gai Z. Rare de novo deletion of metabotropic glutamate receptor 7 (GRM7) gene in a patient with autism spectrum disorder. Am J Med Genet B Neuropsychiatr Genet 2015; 168B:258-64. [PMID: 25921429 DOI: 10.1002/ajmg.b.32306] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 02/22/2015] [Indexed: 12/28/2022]
Abstract
GRM7, the gene encoding metabotropic glutamate receptor 7 (mGluR7), have been implicated in multiple neuropsychiatric disorders and shown to mediate excitatory synaptic neurotransmitter signaling and plasticity in the mammalian brain. Here we report a 303 kb de novo deletion at band 3p26.1, disrupting five coding exons of GRM7 in a proband with autism spectrum disorder, and hyperactivity. Our exon transcriptome-mutation contingency index method shows that three of the exons within the breakpoint boundaries are under purifying selection and highly expressed in prenatal brain regions. Based on our results and a thorough review of the literature, we propose that haploinsufficiency of the GRM7 product (mGluR7) contributes to autism spectrum disorders and hyperactivity phenotype as seen in the patient described here.
Collapse
Affiliation(s)
- Yi Liu
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Yanqing Zhang
- Pediatric Health Care Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Dongmei Zhao
- Pediatric Health Care Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Rui Dong
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Xiaomeng Yang
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Kristiina Tammimies
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada.,The Center of Neurodevelopmental Disorders at Karolinska Institutet (KIND), Pediatric Neuropsychiatry Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Mohammed Uddin
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada.,McLaughlin Centre and Department of Molecular Genetics, University ofToronto, Toronto, Canada
| | - Zhongtao Gai
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China.,Pediatric Health Care Institute, Qilu Children's Hospital of Shandong University, Ji'nan, China
| |
Collapse
|
46
|
Abstract
The brain is composed of many lipids with varied forms that serve not only as structural components but also as essential signaling molecules. Lysophosphatidic acid (LPA) is an important bioactive lipid species that is part of the lysophospholipid (LP) family. LPA is primarily derived from membrane phospholipids and signals through six cognate G protein-coupled receptors (GPCRs), LPA1-6. These receptors are expressed on most cell types within central and peripheral nervous tissues and have been functionally linked to many neural processes and pathways. This Review covers a current understanding of LPA signaling in the nervous system, with particular focus on the relevance of LPA to both physiological and diseased states.
Collapse
Affiliation(s)
- Yun C Yung
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nicole C Stoddard
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Biomedical Sciences Graduate Program, University of California, San Diego School of Medicine, La Jolla, CA 92037, USA
| | - Hope Mirendil
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jerold Chun
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
47
|
Butler MG, Rafi SK, Manzardo AM. High-resolution chromosome ideogram representation of currently recognized genes for autism spectrum disorders. Int J Mol Sci 2015; 16:6464-95. [PMID: 25803107 PMCID: PMC4394543 DOI: 10.3390/ijms16036464] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 03/11/2015] [Accepted: 03/16/2015] [Indexed: 11/16/2022] Open
Abstract
Recently, autism-related research has focused on the identification of various genes and disturbed pathways causing the genetically heterogeneous group of autism spectrum disorders (ASD). The list of autism-related genes has significantly increased due to better awareness with advances in genetic technology and expanding searchable genomic databases. We compiled a master list of known and clinically relevant autism spectrum disorder genes identified with supporting evidence from peer-reviewed medical literature sources by searching key words related to autism and genetics and from authoritative autism-related public access websites, such as the Simons Foundation Autism Research Institute autism genomic database dedicated to gene discovery and characterization. Our list consists of 792 genes arranged in alphabetical order in tabular form with gene symbols placed on high-resolution human chromosome ideograms, thereby enabling clinical and laboratory geneticists and genetic counsellors to access convenient visual images of the location and distribution of ASD genes. Meaningful correlations of the observed phenotype in patients with suspected/confirmed ASD gene(s) at the chromosome region or breakpoint band site can be made to inform diagnosis and gene-based personalized care and provide genetic counselling for families.
Collapse
Affiliation(s)
- Merlin G Butler
- Departments of Psychiatry & Behavioral Sciences and Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Syed K Rafi
- Departments of Psychiatry & Behavioral Sciences and Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Ann M Manzardo
- Departments of Psychiatry & Behavioral Sciences and Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
48
|
Zantomio D, Chana G, Laskaris L, Testa R, Everall I, Pantelis C, Skafidas E. Convergent evidence for mGluR5 in synaptic and neuroinflammatory pathways implicated in ASD. Neurosci Biobehav Rev 2015; 52:172-7. [PMID: 25704074 DOI: 10.1016/j.neubiorev.2015.02.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/25/2015] [Accepted: 02/07/2015] [Indexed: 01/05/2023]
Abstract
The pathogenesis of Autism Spectrum Disorder (ASD), a serious neurodevelopmental disorder, is poorly understood. We review evidence for alterations in glutamatergic signalling in the aetiology of ASD, with a focus on the metabotropic glutamate receptor-5 (mGluR5). mGluR5 signalling is important for synapse formation, neuroplasticity and long term potentiation as well as neuroprotection and has been shown to have a regulatory role in neuroinflammation. Evidence for neuroinflammation in ASD is supported by increase in pro-inflammatory cytokines in the blood and cerebrospinal fluid (CSF) and increased number and activation of microglia in postmortem dorsolateral prefrontal cortex (DLPFC). mGlur5 signalling has also been shown to downregulate microglial activation. Therefore, we focus on mGluR5 as a potential unifying explanation for synapse alteration and neuroinflammation seen in ASD. Data from mGluR5 knockout mouse models, and syndromic and non syndromic forms of ASD are discussed in relation to how alterations in mGluR5 are associated with ASD symptoms. This review supports altered mGluR5 functioning as a convergent point in ASD pathogenesis and indicates more research is warranted into mGluR5 as a potential therapeutic target.
Collapse
Affiliation(s)
- Daniela Zantomio
- Department of Haematology, Austin Health, Heidelberg, VIC, Australia; Centre for Neural Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Gursharan Chana
- Centre for Neural Engineering, The University of Melbourne, Parkville, Victoria, Australia; Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, Australia
| | - Liliana Laskaris
- Centre for Neural Engineering, The University of Melbourne, Parkville, Victoria, Australia; Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia
| | - Renee Testa
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Parkville, Victoria, Australia; Department of Psychology, Monash University, Clayton, Vic, Australia
| | - Ian Everall
- Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, Australia
| | - Christos Pantelis
- Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia; Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Parkville, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, Australia
| | - Efstratios Skafidas
- Centre for Neural Engineering, The University of Melbourne, Parkville, Victoria, Australia; Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, Australia; Centre for Integrative Brain Function, Australia.
| |
Collapse
|
49
|
Abstract
In recent years, considerable progress has been made in understanding the genomic basis of autism spectrum disorder (ASD). Hundreds of variants have been proposed as predisposing to ASD, and the challenge now is to validate candidates and to understand how gene networks interact to produce ASD phenotypes. Genome-wide association and second-generation sequencing studies in particular have provided important indications about how to understand ASD on a molecular level, and we are beginning to see these experimental approaches translate into novel treatments and diagnostic tests. We review key studies in the field over the past five years and discuss some of the remaining technological and methodological challenges that remain.
Collapse
|
50
|
|