1
|
Ledbetter DH, Finucane B, Moreno-De-Luca D, Myers SM. Mainstreaming Diagnostic Genetic Testing and Precision Medicine for Autism Spectrum Disorder: The Role of Child and Adolescent Psychiatrists. Psychiatr Clin North Am 2025; 48:343-360. [PMID: 40348422 DOI: 10.1016/j.psc.2025.01.010] [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] [Indexed: 05/14/2025]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental psychiatric condition that shares significant clinical and genetic overlap with intellectualdisability (ID) and other neurodevelopmental disorders. Genetic testingin ASD lags far behind that for ID, even though Professional Societiesrecommend genetic testing for all ASD individuals and insurance reimbursement is relatively good. The core competencies for child and adolescent psychiatrists include determining the etiology and diagnosisfor all childhood psychopathology, including ID and ASD. Child psychiatrists should recommend and order genetic testing by exomeor genome sequencing on all children with ASD.
Collapse
Affiliation(s)
- David H Ledbetter
- Department of Clinical Sciences, Institute for Pediatric Rare Diseases, Florida State University, 1115 West Call Street, Tallahassee, FL 32306, USA.
| | - Brenda Finucane
- Department of Developmental Medicine, Geisinger College of Health Sciences, 120 Hamm Drive, Lewisburg, PA 17837, USA
| | - Daniel Moreno-De-Luca
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; Precision Medicine in Autism (PRISMA) Group; University of Alberta, Alberta Health Services, CASA Mental Health, 11361 87 Avenue, Suite 5-020K, Edmonton, AB T6G 2E1, Canada
| | - Scott M Myers
- Department of Developmental Medicine, Geisinger College of Health Sciences, 120 Hamm Drive, Lewisburg, PA 17837, USA
| |
Collapse
|
2
|
Kim AY, Yehia L, Eng C. Genomic diversity in functionally relevant genes modifies neurodevelopmental versus neoplastic risks in individuals with germline PTEN variants. NPJ Genom Med 2025; 10:43. [PMID: 40394016 PMCID: PMC12092801 DOI: 10.1038/s41525-025-00495-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/24/2025] [Indexed: 05/22/2025] Open
Abstract
Individuals with germline PTEN variants (PHTS) have increased risks of the seemingly disparate phenotypes of cancer and neurodevelopmental disorders (NDD), including autism spectrum disorder (ASD). Etiology of the phenotypic variability remains elusive. Here, we hypothesized that decreased genomic diversity, manifested by increased homozygosity, may be one etiology. Comprehensive analyses of 376 PHTS patients of European ancestry revealed significant enrichment of homozygous common variants in genes involved in inflammatory processes in the PHTS-NDD group and in genes involved in differentiation and chromatin structure regulation in the PHTS-ASD group. Pathway analysis revealed pathways germane to NDD/ASD, including neuroinflammation and synaptogenesis. Collapsing analysis of the homozygous variants identified suggestive modifier NDD/ASD genes. In contrast, we found enrichment of homozygous ultra-rare variants in genes modulating cell death in the PHTS-cancer group. Finally, homozygosity burden as a predictor of ASD versus cancer outcomes in our validated prediction model for NDD/ASD performed favorably.
Collapse
Affiliation(s)
- Adriel Y Kim
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Lamis Yehia
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Charis Eng
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Center for Personalized Genetic Healthcare, Medical Specialties Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Germline High Risk Cancer Focus Group, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
3
|
Rynard KM, Han K, Wainberg M, Calarco JA, Lee HO, Lipshitz HD, Smibert CA, Tripathy SJ. ASiDentify (ASiD): a machine learning model to predict new autism spectrum disorder risk genes. Genetics 2025; 230:iyaf040. [PMID: 40088463 DOI: 10.1093/genetics/iyaf040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that affects nearly 3% of children and has a strong genetic component. While hundreds of ASD risk genes have been identified through sequencing studies, the genetic heterogeneity of ASD makes identifying additional risk genes using these methods challenging. To predict candidate ASD risk genes, we developed a simple machine learning model, ASiDentify (ASiD), using human genomic, RNA- and protein-based features. ASiD identified over 1,300 candidate ASD risk genes, over 300 of which have not been previously predicted. ASiD made accurate predictions of ASD risk genes using 6 features predictive of ASD risk gene status, including mutational constraint, synapse localization and gene expression in neurons, astrocytes and non-brain tissues. Particular functional groups of proteins found to be strongly implicated in ASD include RNA-binding proteins (RBPs) and chromatin regulators. We constructed additional logistic regression models to make predictions and assess informative features specific to RBPs, including mutational constraint, or chromatin regulators, for which both expression level in excitatory neurons and mutational constraint were informative. The fact that RBPs and chromatin regulators had informative features distinct from all protein-coding genes suggests that specific biological pathways connect risk genes with different molecular functions to ASD.
Collapse
Affiliation(s)
- Katherine M Rynard
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kara Han
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Krembil Institute for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Michael Wainberg
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Krembil Institute for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - John A Calarco
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Hyun O Lee
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Howard D Lipshitz
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Craig A Smibert
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shreejoy J Tripathy
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Krembil Institute for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
4
|
Hiramoto T, Sumiyoshi A, Kato R, Yamauchi T, Takano T, Kang G, Esparza M, Matsumura B, Stevens LJ, Hiroi YJ, Tanifuji T, Ryoke R, Nonaka H, Machida A, Nomoto K, Mogi K, Kikusui T, Kawashima R, Hiroi N. Highly demarcated structural alterations in the brain and impaired social incentive learning in Tbx1 heterozygous mice. Mol Psychiatry 2025; 30:1876-1886. [PMID: 39463450 PMCID: PMC12014486 DOI: 10.1038/s41380-024-02797-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024]
Abstract
Copy number variants (CNVs) are robustly associated with psychiatric disorders and changes in brain structures. However, because CNVs contain many genes, the precise gene-phenotype relationship remains unclear. Although various volumetric alterations in the brains of 22q11.2 CNV carriers have been identified in humans and mouse models, it is unknown how each gene encoded in the 22q11.2 region contributes to structural alterations, associated mental illnesses, and their dimensions. Our previous studies identified Tbx1, a T-box family transcription factor encoded in the 22q11.2 CNV, as a driver gene for social interaction and communication, spatial and working memory, and cognitive flexibility. However, it remains unclear how TBX1 impacts the volumes of various brain regions and their functionally linked behavioral dimensions. In this study, we used volumetric magnetic resonance imaging analysis to comprehensively evaluate brain region volumes and behavioral alterations relevant to affected structures in congenic Tbx1 heterozygous mice. Our data showed that the volumes of the anterior and posterior portions of the amygdaloid complex and its surrounding cortical regions were most robustly reduced in Tbx1 heterozygous mice. In an amygdala-dependent task, Tbx1 heterozygous mice were impaired in their ability to learn the incentive value of a social partner. The volumes of the primary and secondary auditory cortexes were increased, and acoustic, but not non-acoustic, sensorimotor gating was impaired in Tbx1 heterozygous mice. Our findings identify the brain's regional volume alterations and their relevant behavioral dimensions associated with Tbx1 heterozygosity.
Collapse
Affiliation(s)
| | - Akira Sumiyoshi
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Risa Kato
- Laboratory of Human-Animal Interaction and Reciprocity, Azabu University, Sagamihara, Kanagawa, Japan
| | | | - Takeshi Takano
- Department of Pharmacology, UT Health, San Antonio, TX, USA
| | - Gina Kang
- Department of Pharmacology, UT Health, San Antonio, TX, USA
| | - Marisa Esparza
- Department of Pharmacology, UT Health, San Antonio, TX, USA
| | | | | | - Yukiko J Hiroi
- Department of Pharmacology, UT Health, San Antonio, TX, USA
| | | | - Rie Ryoke
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hiroi Nonaka
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Akihiro Machida
- Laboratory of Human-Animal Interaction and Reciprocity, Azabu University, Sagamihara, Kanagawa, Japan
| | - Kensaku Nomoto
- Laboratory of Human-Animal Interaction and Reciprocity, Azabu University, Sagamihara, Kanagawa, Japan
| | - Kazutaka Mogi
- Laboratory of Human-Animal Interaction and Reciprocity, Azabu University, Sagamihara, Kanagawa, Japan
| | - Takefumi Kikusui
- Laboratory of Human-Animal Interaction and Reciprocity, Azabu University, Sagamihara, Kanagawa, Japan
| | - Ryuta Kawashima
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Noboru Hiroi
- Department of Pharmacology, UT Health, San Antonio, TX, USA.
- Department of Cellular and Integrative Physiology, UT Health, San Antonio, TX, USA.
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX, USA.
| |
Collapse
|
5
|
Qu W, Yan G, Du Y, Zhou X, Huang C, Li B, Zhou J, Li Q. Crosstalk Between Mitochondrial DNA and Immune Response: Focus on Autism Spectrum Disorder. Mol Neurobiol 2025; 62:5629-5639. [PMID: 39589631 DOI: 10.1007/s12035-024-04637-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/16/2024] [Indexed: 11/27/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by multiple dysfunctions in behavior, the nervous system, and the immune system. Increasing evidence suggests that mitochondrial DNA (mtDNA) plays a crucial role in the pathology of ASD. In clinical practice, altered mtDNA levels have been observed in various tissues of individuals with ASD. Mutation or oxidation of mtDNA is also closely related to the immune response associated with the pathology of autism. With mtDNA identified as a causal factor, much interest has focused on how its production affects neurodevelopment and neurophysiology. Here, we review how mtDNA leads to dysfunction of cellular mitochondria and immune response. We also illustrate the relationship between mtDNA alterations and the pathology of autism. Finally, we discuss the existing evidence on cell-free mtDNA associated with ASD and look forward to its application in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Wenxuan Qu
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Ge Yan
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Yajuan Du
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Xinyang Zhou
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Chutian Huang
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Bei Li
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Junmei Zhou
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Qian Li
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China.
| |
Collapse
|
6
|
Tan S, Zhang Q, Zhan R, Luo S, Han Y, Yu B, Muss C, Pingault V, Marlin S, Delahaye A, Peters S, Perne C, Kreiß M, Spataro N, Trujillo-Quintero JP, Racine C, Tran-Mau-Them F, Phornphutkul C, Besterman AD, Martinez J, Wang X, Tian X, Srivastava S, Urion DK, Madden JA, Saif HA, Morrow MM, Begtrup A, Li X, Jurgensmeyer S, Leahy P, Zhou S, Li F, Hu Z, Tan J, Xia K, Guo H. Monoallelic loss-of-function variants in GSK3B lead to autism and developmental delay. Mol Psychiatry 2025; 30:1952-1965. [PMID: 39472663 DOI: 10.1038/s41380-024-02806-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 10/09/2024] [Accepted: 10/18/2024] [Indexed: 04/24/2025]
Abstract
De novo variants adjacent to the canonical splicing sites or in the well-defined splicing-related regions are more likely to impair splicing but remain under-investigated in autism spectrum disorder (ASD). By analyzing large, recent ASD genome sequencing cohorts, we find a significant burden of de novo potential splicing-disrupting variants (PSDVs) in 5048 probands compared to 4090 unaffected siblings. We identified 55 genes with recurrent de novo PSDVs that were highly intolerant to variation. Forty-six of these genes have not been strongly implicated in ASD or other neurodevelopmental disorders previously, including GSK3B. Through international, multicenter collaborations, we assembled genotype and phenotype data for 15 individuals with GSK3B variants and identified common phenotypes including developmental delay, ASD, sleeping disturbance, and aggressive behavior. Using available single-cell transcriptomic data, we show that GSK3B is enriched in dorsal progenitors and intermediate forms of excitatory neurons in the developing brain. We showed that Gsk3b knockdown in mouse excitatory neurons interferes with dendrite arborization and spine maturation which could not be rescued by de novo missense variants identified from affected individuals. In summary, our findings suggest that PSDVs may play an important role in the genetic etiology of ASD and allow for the prioritization of new ASD candidate genes. Importantly, we show that genetic variation resulting in GSK3B loss-of-function can lead to a neurodevelopmental disorder with core features of ASD and developmental delay.
Collapse
Affiliation(s)
- Senwei Tan
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Qiumeng Zhang
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Rui Zhan
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Si Luo
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yaoling Han
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Bin Yu
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Candace Muss
- Department of Genetics, Nemours Children's Hospital, Wilmington, DE, USA
| | - Veronique Pingault
- Service de Médecine Génomique des maladies rares, AP-HP, Hôpital Necker; Université Paris Cité, Inserm, Institut Imagine; and Laboratoire de Biologie Médicale Multi-Sites SeqOIA, Paris, France
| | - Sandrine Marlin
- Centre de Référence «Surdités Génétiques», Fédération de Génétique; Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
- Laboratory of Embryology and Genetics of Malformations, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Andrée Delahaye
- Service de Médecine Génomique des maladies rares, AP-HP, Hôpital Necker; Université Paris Cité, Inserm, Institut Imagine; and Laboratoire de Biologie Médicale Multi-Sites SeqOIA, Paris, France
| | - Sophia Peters
- Institute of Human Genetics, School of Medicine, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Claudia Perne
- Institute of Human Genetics, School of Medicine, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Martina Kreiß
- Institute of Human Genetics, School of Medicine, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Nino Spataro
- Center for Genomic Medicine, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Juan Pablo Trujillo-Quintero
- Center for Genomic Medicine, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Caroline Racine
- Unité Fonctionnelle d'Innovation diagnostique des maladies rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Frederic Tran-Mau-Them
- Unité Fonctionnelle d'Innovation diagnostique des maladies rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Chanika Phornphutkul
- Division of Human Genetics, Department of Pediatrics, Warren Alpert Medical School of Brown University, Hasbro Children's Hospital, Providence, RI, USA
| | - Aaron D Besterman
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, USA
- Rady Children's Hospital, San Diego, CA, USA
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
| | - Julian Martinez
- Departments of Human Genetics, Pediatrics and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Xiuxia Wang
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoyu Tian
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Siddharth Srivastava
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, MA, USA
| | - David K Urion
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Jill A Madden
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Hind Al Saif
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Virginia Commonwealth, Richmond, VA, USA
| | | | | | - Xing Li
- Departments of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Sarah Jurgensmeyer
- Division of Genetics, Genomics and Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Peter Leahy
- Division of Genetics, Genomics and Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Shimin Zhou
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Faxiang Li
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Zhengmao Hu
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Jieqiong Tan
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Kun Xia
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China.
- MOE Key Lab of Rare Pediatric Diseases, School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Hui Guo
- Center for Medical Genetics & MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China.
- Furong Laboratory, Changsha, Hunan, China.
| |
Collapse
|
7
|
Sznajder ŁJ, Khan M, Ciesiołka A, Tadross M, Nutter CA, Taylor K, Pearson CE, Lewis MH, Hines RM, Swanson MS, Sobczak K, Yuen RKC. Autism-related traits in myotonic dystrophy type 1 model mice are due to MBNL sequestration and RNA mis-splicing of autism-risk genes. Nat Neurosci 2025:10.1038/s41593-025-01943-0. [PMID: 40259070 DOI: 10.1038/s41593-025-01943-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 03/14/2025] [Indexed: 04/23/2025]
Abstract
Genome-wide enrichment of gene-specific tandem repeat expansions has been linked to autism spectrum disorder. One such mutation is the CTG tandem repeat expansion in the 3' untranslated region of the DMPK gene, which is known to cause myotonic muscular dystrophy type 1. Although there is a clear clinical association between autism and myotonic dystrophy, the molecular basis for this connection remains unknown. Here, we report that sequestration of MBNL splicing factors by mutant DMPK RNAs with expanded CUG repeats alters the RNA splicing patterns of autism-risk genes during brain development, particularly a class of autism-relevant microexons. We demonstrate that both DMPK-CTG expansion and Mbnl null mouse models recapitulate autism-relevant mis-splicing profiles, along with social behavioral deficits and altered responses to novelty. These findings support our model that myotonic dystrophy-associated autism arises from developmental mis-splicing of autism-risk genes.
Collapse
Affiliation(s)
- Łukasz J Sznajder
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, NV, USA.
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA.
| | - Mahreen Khan
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Adam Ciesiołka
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznań, Poland
| | - Mariam Tadross
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA
- Department of Psychiatry, McKnight Brain Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Curtis A Nutter
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Katarzyna Taylor
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznań, Poland
| | - Christopher E Pearson
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mark H Lewis
- Department of Psychiatry, McKnight Brain Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Rochelle M Hines
- Department of Psychology, University of Nevada, Las Vegas, NV, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Krzysztof Sobczak
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznań, Poland
| | - Ryan K C Yuen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Shil A, Arava N, Levi N, Levine L, Golan H, Meiri G, Michaelovski A, Tsadaka Y, Aran A, Menashe I. An integrative scoring approach for prioritization of rare autism spectrum disorder candidate variants from whole exome sequencing data. Sci Rep 2025; 15:13024. [PMID: 40234546 PMCID: PMC12000308 DOI: 10.1038/s41598-025-96063-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 03/25/2025] [Indexed: 04/17/2025] Open
Abstract
Discerning clinically relevant autism spectrum disorder (ASD) candidate variants from whole-exome sequencing (WES) data is complex, time-consuming, and labor-intensive. To this end, we developed AutScore, an integrative prioritization algorithm of ASD candidate variants from WES data and assessed its performance to detect clinically relevant variants. We studied WES data from 581 ASD probands, and their parents registered in the Azrieli National Center database for Autism and Neurodevelopment Research. We focused on rare allele frequency (< 1%) and high-quality proband-specific variants affecting genes associated with ASD or other neurodevelopmental disorders (NDDs). We developed AutScore and AutScore.r and assigned each variant based on their pathogenicity, clinical relevance, gene-disease association, and inheritance patterns. Finally, we compared the performance of both AutScore versions with the rating of clinical experts and the NDD variant prioritization algorithm, AutoCaSc. Overall, 1161 rare variants distributed in 687 genes in 441 ASD probands were evaluated by AutScore with scores ranging from - 4 to 25, with a mean ± SD of 5.89 ± 4.18. AutScore.r cut-off of ≥ 0.335 performs better than AutoCaSc and AutScore in detecting clinically relevant ASD variants, with a detection accuracy rate of 85% and an overall diagnostic yield of 10.3%. Five variants with AutScore.r of ≥ 0.335 were distributed in five novel ASD candidate genes. AutScore.r is an effective automated ranking system for ASD candidate variants that could be implemented in ASD clinical genetics pipelines.
Collapse
Affiliation(s)
- Apurba Shil
- Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Noa Arava
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Noam Levi
- Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Liron Levine
- Bioinformatics Core Facility, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hava Golan
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Gal Meiri
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Preschool Psychiatric Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Analya Michaelovski
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Child Development Center, Soroka University Medical Center, Beer Sheva, Israel
| | - Yair Tsadaka
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Child Development Center, Ministry of Health, Be'er Sheva, 84100, Israel
| | - Adi Aran
- Neuropediatric Unit, Shaare Zedek Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Idan Menashe
- Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
9
|
Tonekaboni SH, Iaboni A, Trost B, Reuter M, Lindenmaier Z, Kushki A, Kelley E, Jones J, Ayub M, Georgiades S, Nicolson R, Chan E, Cretu A, Brian J, Anagnostou E. Comparative Analysis of Phenotypic and Genotypic Differences Between Individuals Affected by Regressive and Non-Regressive Autism: A Cross-Sectional Study. Autism Res 2025. [PMID: 40177835 DOI: 10.1002/aur.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 04/05/2025]
Abstract
Development among autistic youth varies widely. A subgroup of children experiences regression, defined as the loss of previously acquired developmental skills. Various genetic and environmental factors have been suggested as potential contributors. This study aimed to compare the developmental profiles of children and youth with regression to those without and identify factors associated with regression. Data from the Province of Ontario Neurodevelopmental Disorders (POND) Network was analyzed, including 930 eligible participants. Regression classification was based on the Autism Diagnostic Interview-Revised (ADI-R). Differences in demographic information, medical history, mental health, cognitive and adaptive functioning, and molecular genetic findings were examined between individuals with regressive and non-regressive autism. Among participants, 211 (22.7%) had regressive autism. Lower Full-Scale IQ (p corrected = 0.015) and adaptive function (ABAS-2) scores (p corrected = 0.015) were identified in the regressive group. No statistically significant differences in mental health outcomes (measured by the Child Behavior Checklist, CBCL) or socialization and core symptom severity (measured by the Social Communication Questionnaire, SCQ) were found. There were no notable differences in other factors hypothesized to contribute to regression, such as pregnancy duration, family history of autism, caregivers' education levels, or sleep disorders, except for a higher prevalence of epilepsy in the regressive group (p = 0.001). Rare and common genetic features of both groups are described. In conclusion, autistic youth with regression tend to have lower cognitive and adaptive scores and may experience higher epilepsy rates. Further powered studies are needed to explore the genomic architecture of autistic regression.
Collapse
Affiliation(s)
- Seyed Hassan Tonekaboni
- Autism Research Centre, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Alana Iaboni
- Autism Research Centre, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Brett Trost
- Programs in Molecular Medicine and Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Miriam Reuter
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Zsuzsa Lindenmaier
- Autism Research Centre, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Azadeh Kushki
- Autism Research Centre, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | | | - Jessica Jones
- Department of Psychiatry, Queen's University, Kingston, Canada
| | - Muhammed Ayub
- Department of Psychiatry, University College London, London, UK
| | - Stelios Georgiades
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| | | | - Elim Chan
- Autism Research Centre, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Andrada Cretu
- Autism Research Centre, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Jessica Brian
- Autism Research Centre, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Evdokia Anagnostou
- Autism Research Centre, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Bruxel EM, Rovaris DL, Belangero SI, Chavarría-Soley G, Cuellar-Barboza AB, Martínez-Magaña JJ, Nagamatsu ST, Nievergelt CM, Núñez-Ríos DL, Ota VK, Peterson RE, Sloofman LG, Adams AM, Albino E, Alvarado AT, Andrade-Brito D, Arguello-Pascualli PY, Bandeira CE, Bau CHD, Bulik CM, Buxbaum JD, Cappi C, Corral-Frias NS, Corrales A, Corsi-Zuelli F, Crowley JJ, Cupertino RB, da Silva BS, De Almeida SS, De la Hoz JF, Forero DA, Fries GR, Gelernter J, González-Giraldo Y, Grevet EH, Grice DE, Hernández-Garayua A, Hettema JM, Ibáñez A, Ionita-Laza I, Lattig MC, Lima YC, Lin YS, López-León S, Loureiro CM, Martínez-Cerdeño V, Martínez-Levy GA, Melin K, Moreno-De-Luca D, Muniz Carvalho C, Olivares AM, Oliveira VF, Ormond R, Palmer AA, Panzenhagen AC, Passos-Bueno MR, Peng Q, Pérez-Palma E, Prieto ML, Roussos P, Sanchez-Roige S, Santamaría-García H, Shansis FM, Sharp RR, Storch EA, Tavares MEA, Tietz GE, Torres-Hernández BA, Tovo-Rodrigues L, Trelles P, Trujillo-ChiVacuan EM, Velásquez MM, Vera-Urbina F, Voloudakis G, Wegman-Ostrosky T, Zhen-Duan J, Zhou H, Santoro ML, Nicolini H, Atkinson EG, Giusti-Rodríguez P, Montalvo-Ortiz JL. Psychiatric genetics in the diverse landscape of Latin American populations. Nat Genet 2025:10.1038/s41588-025-02127-z. [PMID: 40175716 DOI: 10.1038/s41588-025-02127-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 02/14/2025] [Indexed: 04/04/2025]
Abstract
Psychiatric disorders are highly heritable and polygenic, influenced by environmental factors and often comorbid. Large-scale genome-wide association studies (GWASs) through consortium efforts have identified genetic risk loci and revealed the underlying biology of psychiatric disorders and traits. However, over 85% of psychiatric GWAS participants are of European ancestry, limiting the applicability of these findings to non-European populations. Latin America and the Caribbean, regions marked by diverse genetic admixture, distinct environments and healthcare disparities, remain critically understudied in psychiatric genomics. This threatens access to precision psychiatry, where diversity is crucial for innovation and equity. This Review evaluates the current state and advancements in psychiatric genomics within Latin America and the Caribbean, discusses the prevalence and burden of psychiatric disorders, explores contributions to psychiatric GWASs from these regions and highlights methods that account for genetic diversity. We also identify existing gaps and challenges and propose recommendations to promote equity in psychiatric genomics.
Collapse
Affiliation(s)
- Estela M Bruxel
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Diego L Rovaris
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Sintia I Belangero
- Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
- Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Gabriela Chavarría-Soley
- Escuela de Biología y Centro de Investigación en Biología Celular y Molecular, Universidad de Costa Rica, San Pedro, Costa Rica
| | - Alfredo B Cuellar-Barboza
- Department of Psychiatry, School of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, México
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - José J Martínez-Magaña
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Psychiatry Division, VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Sheila T Nagamatsu
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Psychiatry Division, VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Caroline M Nievergelt
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Diana L Núñez-Ríos
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Psychiatry Division, VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Vanessa K Ota
- Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
- Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Roseann E Peterson
- Department of Psychiatry and Behavioral Sciences, Institute for Genomics in Health, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Laura G Sloofman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amy M Adams
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, College Station, TX, USA
| | - Elinette Albino
- School of Health Professions, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Angel T Alvarado
- Research Unit in Molecular Pharmacology and Genomic Medicine, VRI, San Ignacio de Loyola University, La Molina, Perú
| | | | - Paola Y Arguello-Pascualli
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cibele E Bandeira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Claiton H D Bau
- Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Cynthia M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Joseph D Buxbaum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolina Cappi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Alejo Corrales
- Departamento de Psiquiatría, Universidad Nacional de Tucumán, San Miguel de Tucumán, Argentina
| | - Fabiana Corsi-Zuelli
- Department of Neuroscience, Ribeirão Preto Medical School, Universidade de São Paulo, São Paulo, Brazil
| | - James J Crowley
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Renata B Cupertino
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Bruna S da Silva
- Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Suzannah S De Almeida
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juan F De la Hoz
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Diego A Forero
- School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá, Colombia
| | - Gabriel R Fries
- Faillace Department of Psychiatry and Behavioral Sciences, the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Psychiatry Division, VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Yeimy González-Giraldo
- Biomedical Sciences Research Group, School of Medicine, Universidad Antonio Nariño, Bogotá, Colombia
| | - Eugenio H Grevet
- Department of Psychiatry and Legal Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Dorothy E Grice
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adriana Hernández-Garayua
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Psychiatry Division, VA Connecticut Healthcare Center, West Haven, CT, USA
| | - John M Hettema
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, College Station, TX, USA
| | - Agustín Ibáñez
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Iuliana Ionita-Laza
- Department of Biostatistics, Columbia University, New York, NY, USA
- Department of Statistics, Lund University, Lund, Sweden
| | | | - Yago C Lima
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Yi-Sian Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Sandra López-León
- Quantitative Safety Epidemiology, Novartis Pharma, East Hanover, NJ, USA
- Rutgers Center for Pharmacoepidemiology and Treatment Science, Rutgers University, New Brunswick, NJ, USA
| | - Camila M Loureiro
- Department of Neuroscience, Ribeirão Preto Medical School, Universidade de São Paulo, São Paulo, Brazil
| | | | - Gabriela A Martínez-Levy
- Department of Genetics, Subdirectorate of Clinical Research, National Institute of Psychiatry, México City, México
- Department of Cell and Tissular Biology, Medicine Faculty, National Autonomous University of Mexico, México City, México
| | - Kyle Melin
- School of Pharmacy, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Daniel Moreno-De-Luca
- Precision Medicine in Autism Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Alberta Health Services, CASA Mental Health, Edmonton, Alberta, Canada
| | | | - Ana Maria Olivares
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Boston, MA, USA
| | - Victor F Oliveira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Rafaella Ormond
- Disciplina de Biologia Molecular, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alana C Panzenhagen
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden
- Laboratório de Pesquisa Translacional em Comportamento Suicida, Universidade do Vale do Taquari, Lajeado, Brazil
| | - Maria Rita Passos-Bueno
- Departmento de Genetica e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Qian Peng
- Department of Neuroscience, the Scripps Research Institute, La Jolla, CA, USA
| | - Eduardo Pérez-Palma
- Facultad de Medicina Clínica Alemana, Centro de Genética y Genómica, Universidad del Desarrollo, Santiago, Chile
| | - Miguel L Prieto
- Mental Health Service, Clínica Universidad de los Andes, Santiago, Chile
- Department of Psychiatry, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Panos Roussos
- Center for Disease Neurogenomics, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hernando Santamaría-García
- PhD Program of Neuroscience, Pontificia Universidad Javeriana, Hospital San Ignacio, Center for Memory and Cognition, Intellectus, Bogotá, Colombia
| | - Flávio M Shansis
- Graduate Program of Medical Sciences, Universidade do Vale do Taquari, Lajeado, Brazil
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Rachel R Sharp
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eric A Storch
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Maria Eduarda A Tavares
- Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Grace E Tietz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Pilar Trelles
- Department of Psychiatry and Behavioral Sciences, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eva M Trujillo-ChiVacuan
- Research Department, Comenzar de Nuevo Eating Disorders Treatment Center, Monterrey, México
- Escuela de Medicina y Ciencias de la Salud Tecnológico de Monterrey, Monterrey, México
| | - Maria M Velásquez
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Fernando Vera-Urbina
- School of Pharmacy, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Georgios Voloudakis
- Center for Disease Neurogenomics, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Jenny Zhen-Duan
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Hang Zhou
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Psychiatry Division, VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Marcos L Santoro
- Disciplina de Biologia Molecular, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Humberto Nicolini
- Laboratorio de Enfermedades Psiquiátricas, Neurodegenerativas y Adicciones, Instituto Nacional de Medicina Genómica, Mexico City, México
| | - Elizabeth G Atkinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Center, Texas Children's Hospital, Houston, TX, USA.
| | - Paola Giusti-Rodríguez
- Department of Psychiatry, University of Florida College of Medicine, Gainesville, FL, USA.
| | - Janitza L Montalvo-Ortiz
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Psychiatry Division, VA Connecticut Healthcare Center, West Haven, CT, USA.
- Department of Biomedical Informatics and Data Science, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
11
|
Gao S, Shan C, Zhang R, Wang T. Genetic advances in neurodevelopmental disorders. MEDICAL REVIEW (2021) 2025; 5:139-151. [PMID: 40224365 PMCID: PMC11987507 DOI: 10.1515/mr-2024-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/14/2024] [Indexed: 04/15/2025]
Abstract
Neurodevelopmental disorders (NDDs) are a group of highly heterogeneous diseases that affect children's social, cognitive, and emotional functioning. The etiology is complicated with genetic factors playing an important role. During the past decade, large-scale whole exome sequencing (WES) and whole genome sequencing (WGS) have vastly advanced the genetic findings of NDDs. Various forms of variants have been reported to contribute to NDDs, such as de novo mutations (DNMs), copy number variations (CNVs), rare inherited variants (RIVs), and common variation. By far, over 200 high-risk NDD genes have been identified, which are involved in biological processes including synaptic function, transcriptional and epigenetic regulation. In addition, monogenic, oligogenic, polygenetic, and omnigenic models have been proposed to explain the genetic architecture of NDDs. However, the majority of NDD patients still do not have a definitive genetic diagnosis. In the future, more types of risk factors, as well as noncoding variants, are await to be identified, and including their interplay mechanisms are key to resolving the etiology and heterogeneity of NDDs.
Collapse
Affiliation(s)
- Shilin Gao
- Department of Neuroscience, Neuroscience Research Institute, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, China
| | - Chaoyi Shan
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Rong Zhang
- Department of Neuroscience, Neuroscience Research Institute, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, China
| | - Tianyun Wang
- Department of Neuroscience, Neuroscience Research Institute, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, China
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Autism Research Center, Peking University Health Science Center, Beijing, China
| |
Collapse
|
12
|
Selvanayagam T, Hoang N, Sarikaya E, Howe J, Russell C, Iaboni A, Quirbach M, Marshall CR, Szatmari P, Anagnostou E, Vorstman J, Hartley DM, Scherer SW. Clinical utility of genome sequencing in autism: illustrative examples from a genomic research study. J Med Genet 2025:jmg-2024-110463. [PMID: 40169255 DOI: 10.1136/jmg-2024-110463] [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: 10/25/2024] [Accepted: 03/04/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Genetics is an important contributor to autism spectrum disorder (ASD). Clinical guidelines endorse genetic testing in the medical workup of ASD, particularly tests that use whole genome sequencing (WGS) technology. While the clinical utility of genetic testing in ASD is demonstrated, the breadth of impact of results can depend on the variant and/or gene being reported. METHODS We reviewed research results returned to families enrolled in our ASD WGS study between 2012 and 2023. For significant results, we grouped the outcome of each genetic finding into three outcome categories: (1) genetic diagnosis, (2) counselling benefits and (3) support to family. RESULTS Out of 202 families who received genome sequencing results, 100 had at least one clinically relevant finding related to ASD. With detailed examples, we show that all significant results led to a genetic diagnosis and counselling benefits. CONCLUSION Our findings show the relevance of genome sequencing in ASD and provide illustrative examples of how the information can be used.
Collapse
Affiliation(s)
- Thanuja Selvanayagam
- Department of Genetic Counselling, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, SickKids Research Institute, Toronto, Ontario, Canada
- Autism Research Unit, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ny Hoang
- Department of Genetic Counselling, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, SickKids Research Institute, Toronto, Ontario, Canada
- Autism Research Unit, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ege Sarikaya
- Department of Genetic Counselling, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, SickKids Research Institute, Toronto, Ontario, Canada
- Autism Research Unit, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jennifer Howe
- Program in Genetics and Genome Biology, SickKids Research Institute, Toronto, Ontario, Canada
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Carolyn Russell
- Offord Centre for Child Studies, McMaster University, Hamilton, Ontario, Canada
| | - Alana Iaboni
- Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | | | - Christian R Marshall
- Genome Diagnostics, Department of Paediatric Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Peter Szatmari
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Evdokia Anagnostou
- Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jacob Vorstman
- Program in Genetics and Genome Biology, SickKids Research Institute, Toronto, Ontario, Canada
- Autism Research Unit, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Stephen W Scherer
- Program in Genetics and Genome Biology, SickKids Research Institute, Toronto, Ontario, Canada
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- McLaughlin Centre, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Metcalf K. Categorical misalignment: Making autism(s) in big data biobanking. SOCIAL STUDIES OF SCIENCE 2025; 55:209-237. [PMID: 39370865 PMCID: PMC11986076 DOI: 10.1177/03063127241288223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The opaque relationship between biology and behavior is an intractable problem for psychiatry, and it increasingly challenges longstanding diagnostic categorizations. While various big data sciences have been repeatedly deployed as potential solutions, they have so far complicated more than they have managed to disentangle. Attending to categorical misalignment, this article proposes one reason why this is the case: Datasets have to instantiate clinical categories in order to make biological sense of them, and they do so in different ways. Here, I use mixed methods to examine the role of the reuse of big data in recent genomic research on autism spectrum disorder (ASD). I show how divergent regimes of psychiatric categorization are innately encoded within commonly used datasets from MSSNG and 23andMe, contributing to a rippling disjuncture in the accounts of autism that this body of research has produced. Beyond the specific complications this dynamic introduces for the category of autism, this paper argues for the necessity of critical attention to the role of dataset reuse and recombination across human genomics and beyond.
Collapse
|
14
|
Besterman AD, Alnor MA, Castaño M, DeLisi LE, Grice DE, Lohoff FW, Middeldorp CM, Müller DJ, Quattrone D, Nurnberger J, Nurmi EL, Ross DA, Soda T, Schulze TG, Trost B, Vilella E, Yap CX, Zai G, Moreno-De-Luca D. Psychiatric Genetics in Clinical Practice: Essential Knowledge for Mental Health Professionals. Am J Psychiatry 2025:appiajp20240295. [PMID: 40134266 DOI: 10.1176/appi.ajp.20240295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
OBJECTIVE The authors provide recommendations on incorporating recent advances in psychiatric genetics into clinical practice for mental health clinicians. METHOD The International Society for Psychiatric Genetics Education Committee met monthly to come to a consensus on priority topics in psychiatric genetics. Topics were then assigned to small teams of subspecialty experts to summarize the current knowledge base and create an illustrative clinical case. Topics included, familial aggregation, common and rare genetic variants, epigenetics, gene-environment interactions, pharmacogenomics, genetic counseling, and ethical and social implications. Each section was reviewed and revised by all committee members and then finalized by the Committee Chair. RESULTS Key findings highlight the importance of understanding the genetic architecture of psychiatric disorders, the potential applications of genetic information in risk assessment, diagnosis, treatment selection, and patient education, as well as the ethical and social considerations surrounding the use of genetic data. The committee emphasizes the need for a nuanced approach that integrates genetic factors with environmental and experiential factors in a holistic model of care. CONCLUSION As psychiatric genetics continues to evolve rapidly, mental health clinicians must stay informed about the latest findings and their clinical implications. Ongoing education, collaboration with genetics professionals, and effective communication strategies are crucial to harness the power of genetics while avoiding potential pitfalls such as genetic determinism and stigma. The committee recommends a balanced perspective that recognizes the complex interplay of genetic and non-genetic factors in shaping mental health outcomes.
Collapse
Affiliation(s)
- Aaron D Besterman
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Mohamed A Alnor
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Mauricio Castaño
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Lynn E DeLisi
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Dorothy E Grice
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Falk W Lohoff
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Christel M Middeldorp
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Daniel J Müller
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Diego Quattrone
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - John Nurnberger
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Erika L Nurmi
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - David A Ross
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Takahiro Soda
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Thomas G Schulze
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Brett Trost
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Elisabet Vilella
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Chloe X Yap
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Gwyneth Zai
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Daniel Moreno-De-Luca
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| |
Collapse
|
15
|
Meert L, Pelicano de Almeida M, Dekker MR, Dekkers DHW, Nowosad K, Huylebroeck D, van den Hout M, Ozgür Z, van IJcken WFJ, Demmers J, Fornerod M, Poot RA. A CHD8-TRRAP axis facilitates MYC and E2F target gene regulation in human neural stem cells. iScience 2025; 28:111978. [PMID: 40104050 PMCID: PMC11914185 DOI: 10.1016/j.isci.2025.111978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/06/2024] [Accepted: 02/05/2025] [Indexed: 03/20/2025] Open
Abstract
Mutations in ATP-dependent chromatin remodeler CHD8 cause one of the most frequent monogenetic forms of autism and are associated with brain overgrowth. Nevertheless, the activities of CHD8 in autism-relevant cell types are still poorly understood. Here, we purify the CHD8 protein from human neural stem cells and determine its interaction partners using mass spectrometry. We identify the TRRAP complex, a coactivator of MYC and E2F transcription factors, as a prominent CHD8 interaction partner. CHD8 colocalizes genome-wide with TRRAP and binds together at MYC and E2F target gene promoters in human neural stem cells. Depletion of CHD8 or TRRAP in human neural stem cells shows downregulation of MYC and E2F target genes as the most prominent gene-regulatory events. Depletion of CHD8 diminishes cell-cycle entry into S-phase. MYC and E2F transcription factors are established oncogenes and regulate cell growth. Our results link CHD8 to TRRAP in facilitating the regulation of MYC and E2F target genes in human neural stem cells.
Collapse
Affiliation(s)
- Lize Meert
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | | | - Mike R Dekker
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - Dick H W Dekkers
- Center for Proteomics, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Karol Nowosad
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | | | - Zeliha Ozgür
- Center for Biomics, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Wilfred F J van IJcken
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
- Center for Biomics, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Jeroen Demmers
- Center for Proteomics, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Maarten Fornerod
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - Raymond A Poot
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| |
Collapse
|
16
|
Li H, Chen Y, Qiu Y. Oxytocin lipidation expanding therapeutics for long-term reversal of autistic behaviors in rats. Int J Pharm 2025; 672:125299. [PMID: 39890086 DOI: 10.1016/j.ijpharm.2025.125299] [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: 01/03/2025] [Revised: 01/09/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficits in social interaction and repetitive, stereotyped behaviors. There is no universally effective pharmacological treatment targeting its core symptoms.Oxytocin, an endogenous polypeptide known as the "social hormone", has shown potential in improving emotional recognition and social interactions in individuals with ASD. However, its clinical application has been limited due to its short half-life and poor blood-brain barrier penetration. To address these challenges, we utilized peptide lipidation technology to enhance the pharmacokinetic properties and brain bioavailability of oxytocin. A series of lipidated oxytocin analogs was designed and synthesized, exhibiting superior brain distribution and pharmacokinetic profiles in valproic acid-induced autistic rat models compared to unmodified oxytocin. Among theseanalogs, C16-modified oxytocin (C16-OT), administered intrathecally, achieved the most extensive brain distribution with limited presence in the blood, resulting in long-lasting improvements in autistic behaviors. These improvements, including enhanced social behaviors and reduced stereotypical actions, were sustained for up to 42 days, contrasting with the brief effects typically reported in previous studies. Furthermore, a comparison of administration routes revealed that intrathecal injection achieved higher brain concentrations and more prolonged social behavioral improvements than intranasal delivery. These findings provide robust preclinical evidence that C16-OT, through optimized lipidation and intrathecal delivery, offers sustained central nervous system activity and significant, long-term reversal of social behavioral deficits in rats with autism.
Collapse
Affiliation(s)
- Honglin Li
- Department of Social Psychology, School of Sociology, Nankai University, Tianjin, China
| | - Ya Chen
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to UESTC, No.55, Block 4, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Yue Qiu
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to UESTC, No.55, Block 4, Southern Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
17
|
Tamada K, Takumi T. Neurodevelopmental impact of CNV models in ASD: Recent advances and future directions. Curr Opin Neurobiol 2025; 92:103001. [PMID: 40090136 DOI: 10.1016/j.conb.2025.103001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/18/2025]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social communication impairments and restricted, repetitive behaviors. ASD exhibits a strong genetic basis, with rare and common genetic variants contributing to its etiology. Copy number variations (CNVs), deletions or duplications of chromosomal segments, have emerged as key contributors to ASD risk. Rare CNVs often demonstrate large effect sizes and can directly cause ASD, while common variants collectively exert subtle influences. Recent advances have identified numerous ASD-associated CNVs, including recurrent loci such as 1q21.1, 2p16.3, 7q11.23, 15q11.2, 15q11-q13, 16p11.2 and 22q11.2. Mouse models carrying these CNVs have provided profound insights into the underlying neurobiological mechanisms. Recent studies integrating transcriptomic, proteomic, and functional imaging approaches have revealed alterations in synaptic function, neuronal differentiation, myelination, metabolic pathways, and circuit connectivity. Notably, investigations leveraging conditional knockout models, high magnetic field MRI, and single-cell analyses highlight disruptions in excitatory-inhibitory balance, white matter integrity, and dynamic gene regulatory networks. Parallel human-based approaches, including iPSC-derived neurons, cerebral organoids, and large-scale single-nucleus sequencing, are combined with animal model data. These integrative strategies promise to refine our understanding of ASD's genetic architecture, bridging the gap between fundamental discoveries in model organisms and clinically relevant biomarkers, subtypes, and therapeutic targets in humans. This review summarizes key findings from recent CNV mouse model studies and highlights emerging technologies applied to human ASD samples. Finally, we outline prospects for translating findings from mouse studies to humans. By illuminating both unique and convergent genetic mechanisms, these advances offer a critical framework for unraveling etiological complexity in ASD.
Collapse
Affiliation(s)
- Kota Tamada
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, Kobe 650-0017, Japan.
| | - Toru Takumi
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, Kobe 650-0017, Japan.
| |
Collapse
|
18
|
Sotgiu S, Cavassa V, Puci MV, Sotgiu MA, Turilli D, Jacono AL, Nuvoli A, Masala S, Barisano G, Carta A. Enlarged perivascular spaces under the dorso-lateral prefrontal cortex and severity of autism. Sci Rep 2025; 15:8142. [PMID: 40059247 PMCID: PMC11891330 DOI: 10.1038/s41598-025-92913-w] [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] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 03/03/2025] [Indexed: 05/13/2025] Open
Abstract
The glymphatic system allows cerebrospinal fluid (CSF) flow along the brain's perivascular spaces (PVS), aiding in the removal of harmful substances into the venous system. Previous studies have suggested that younger males with severe autism spectrum disorder (ASD) exhibit enlarged PVS (ePVS), although the specific regions or extent of PVS enlargement remain unclear. Additionally, it is still unknown whether the localization of ePVS correlates with specific ASD symptoms. Using automated MRI-based PVS quantification, we conducted a descriptive observational study to map the number, diameter, and volume of PVS across 72 brain areas, correlating these features with the clinical severity of autism and the presence or absence of three distinct ASD symptoms: language impairment, stereotypies, and hypersensoriality. The study involved 36 children with ASD (ages 1-9 years). A thorough analysis of ePVS in the white matter underlying 72 cortical areas revealed that different ASD symptoms exhibit specific ePVS localizations. Moreover, ePVS in the white matter beneath the two rostral middle frontal regions were associated with the overall clinical severity of ASD as well as specific symptoms (verbal dysfunction, stereotypies, and hypersensoriality). When these prefrontal subregions experience excessive PVS enlargement, it may lead to hypoactivity in the dorsolateral prefrontal cortex (DLPFC), contributing to the manifestation of ASD symptoms.
Collapse
Affiliation(s)
- Stefano Sotgiu
- Division of Child Neuropsychiatry, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy.
- Division of Child Neuropsychiatry, Azienda Ospedaliero-Universitaria di Sassari, Sassari, Italy.
- Department of Medicine, Surgery and Pharmacy , University of Sassari , Sassari, Italy.
| | - Vanna Cavassa
- Division of Child Neuropsychiatry, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Mariangela Valentina Puci
- Unit of Statistics, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | | | - Davide Turilli
- Radiology Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Alessandro Lo Jacono
- Faculty of Science, Master of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Angela Nuvoli
- Division of Child Neuropsychiatry, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Salvatore Masala
- Radiology Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | | | - Alessandra Carta
- Division of Child Neuropsychiatry, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| |
Collapse
|
19
|
Costa CIS, Madanelo L, Wang JYT, da Silva Campos G, De Sanctis Girardi AC, Scliar M, Monfardini F, de Cássia Mingroni Pavanello R, Cória VR, Vibranovski MD, Krepischi AC, Lourenço NCV, Zatz M, Yamamoto GL, Zachi EC, Passos-Bueno MR. Understanding rare variant contributions to autism: lessons from dystrophin-deficient model. NPJ Genom Med 2025; 10:18. [PMID: 40050609 PMCID: PMC11885547 DOI: 10.1038/s41525-025-00469-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025] Open
Abstract
Duchenne and Becker Muscular Dystrophy are dystrophinopathies with a prevalence of 1:5000-6000 males, caused by pathogenic variants in DMD. These conditions are often accompanied by neurodevelopmental disorders (NDDs) like autism (ASD; ~20%) and intellectual disability (ID; ~30%). However, their low penetrance in dystrophinopathies suggests additional contributing factors. In our study, 83 individuals with dystrophinopathies were clinically evaluated and categorized based on ASD (36 individuals), ID risk (12 individuals), or controls (35 individuals). Exome sequencing analysis revealed an enrichment of risk de novo variants (DNVs) in ASD-DMD individuals (adjusted p value = 0.0356), with the number of DNVs correlating with paternal age (p value = 0.0133). Additionally, DMD-ASD individuals showed a higher average of rare risk variants (RRVs) compared to DMD-Controls (adjusted p value = 0.0285). Gene ontology analysis revealed an enrichment of extracellular matrix-related genes, especially collagens, and Ehlers-Danlos syndrome genes in ASD-DMD and DMD-ID groups. These findings support an oligogenic model for ASD in dystrophinopathies, highlighting the importance of investigating homogenized samples to elucidate ASD's genetic architecture.
Collapse
Affiliation(s)
- Claudia Ismania Samogy Costa
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Luciana Madanelo
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Jaqueline Yu Ting Wang
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Gabriele da Silva Campos
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Ana Cristina De Sanctis Girardi
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Marília Scliar
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Frederico Monfardini
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Rita de Cássia Mingroni Pavanello
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Vivian Romanholi Cória
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Maria Dulcetti Vibranovski
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Ana Cristina Krepischi
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Naila Cristina Vilaça Lourenço
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Mayana Zatz
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Guilherme Lopes Yamamoto
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Elaine Cristina Zachi
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Maria Rita Passos-Bueno
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil.
| |
Collapse
|
20
|
Pagani M, Zerbi V, Gini S, Alvino F, Banerjee A, Barberis A, Basson MA, Bozzi Y, Galbusera A, Ellegood J, Fagiolini M, Lerch J, Matteoli M, Montani C, Pozzi D, Provenzano G, Scattoni ML, Wenderoth N, Xu T, Lombardo M, Milham MP, Martino AD, Gozzi A. Biological subtyping of autism via cross-species fMRI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641400. [PMID: 40093106 PMCID: PMC11908180 DOI: 10.1101/2025.03.04.641400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
It is frequently assumed that the phenotypic heterogeneity in autism spectrum disorder reflects underlying pathobiological variation. However, direct evidence in support of this hypothesis is lacking. Here, we leverage cross-species functional neuroimaging to examine whether variability in brain functional connectivity reflects distinct biological mechanisms. We find that fMRI connectivity alterations in 20 distinct mouse models of autism (n=549 individual mice) can be clustered into two prominent hypo- and hyperconnectivity subtypes. We show that these connectivity profiles are linked to distinct signaling pathways, with hypoconnectivity being associated with synaptic dysfunction, and hyperconnectivity reflecting transcriptional and immune-related alterations. Extending these findings to humans, we identify analogous hypo- and hyperconnectivity subtypes in a large, multicenter resting state fMRI dataset of n=940 autistic and n=1036 neurotypical individuals. Remarkably, hypo- and hyperconnectivity autism subtypes are replicable across independent cohorts (accounting for 25.1% of all autism data), exhibit distinct functional network architecture, are behaviorally dissociable, and recapitulate synaptic and immune mechanisms identified in corresponding mouse subtypes. Our cross-species investigation, thus, decodes the heterogeneity of fMRI connectivity in autism into distinct pathway-specific etiologies, offering a new empirical framework for targeted subtyping of autism.
Collapse
Affiliation(s)
- Marco Pagani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
- IMT School for Advanced Studies, Lucca, Italy
| | - Valerio Zerbi
- Department of Psychiatry, University of Geneva, Switzerland
- Department of Basic Neurosciences, University of Geneva, Switzerland
| | - Silvia Gini
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
- Center for Mind and Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Filomena Alvino
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | | | - Andrea Barberis
- Synaptic Plasticity of Inhibitory Networks, Istituto Italiano di Tecnologia, Genova, Italy
| | - M. Albert Basson
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Yuri Bozzi
- Center for Mind and Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Jacob Ellegood
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | | | - Jason Lerch
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Michela Matteoli
- Humanitas University, Milan, Italy
- CNR Institute of Neuroscience c/o Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Caterina Montani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Davide Pozzi
- CNR Institute of Neuroscience c/o Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational and Integrative Biology. University of Trento, Trento, Italy
| | - Maria Luisa Scattoni
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy
| | | | - Ting Xu
- Center for Integrative Developing Brain, Child Mind Institute, New York, NY, USA
| | - Michael Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy
| | - Michael P Milham
- Center for the Integrative Developmental Neuroscience, Child Mind Institute, New York, NY, USA
| | | | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| |
Collapse
|
21
|
Kanazashi Y, Usui Y, Iwasaki Y, Sasagawa S, Endo M, Yamaguchi M, Johnson TA, Maejima K, Shiraishi K, Kohno T, Yoshida T, Sugano K, Murakami Y, Kamatani Y, Matsumoto N, Matsuda K, Momozawa Y, Nakagawa H. Cancer and disease profiles for PTEN pathogenic variants in Japanese population. J Hum Genet 2025; 70:135-140. [PMID: 39663357 DOI: 10.1038/s10038-024-01311-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
A germline alteration in the PTEN gene causes a spectrum of disorders conceptualized as PTEN hamartoma tumor syndrome (PHTS), which show high risk of tumor development and a highly variable and complex phenotype. The diagnosis of PHTS is established in a proband by identification of a heterozygous germline PTEN pathogenic variant on molecular genetic testing. In this study, to understand more PTEN-associated clinical phenotype and PHTS in a Japanese population, we extracted 128 germline PTEN rare variants from 113,535 adult Japanese registered in Biobank Japan (BBJ), and categorized 29 pathogenic/likely pathogenic variants in 30 individuals (0.0264%) with ClinVar classifications and ACMG/AMP guideline for PTEN. We examined case-control association in 75,238 patients with various types of cancer and 38,297 non-cancer controls, and identified that PTEN pathogenic variants (PVs) were significantly associated with endometrial cancer (OR = 35.7, P = 9.73E-04) and marginally associated with female breast cancer (OR = 19.5, P = 3.92E-03), especially at young onset and with multiple cancers. We observed that among the 127 disease phenotypes the PTEN PV carriers had uterine fibroid, goiter, ovarian cyst, and epilepsy, which is consistent with PTEN-related phenotypes. We also found that weight/height were significantly higher in adult female carriers with PTEN PV (P = 3.1E-04 and P = 0.0014, respectively), which is consistent with overgrowth syndrome of PHTS. Our results indicate the phenotypical features associated with PTEN PVs in a Japanese population, especially female, and can contribute to the screening for PTEN variants and its associated several phenotypes.
Collapse
Affiliation(s)
- Yuki Kanazashi
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yoshiaki Usui
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yusuke Iwasaki
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shota Sasagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mikiko Endo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mitsuyo Yamaguchi
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Todd A Johnson
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kazuhiro Maejima
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Teruhiko Yoshida
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Tokyo, Japan
| | - Kokichi Sugano
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Tokyo, Japan
- Dapartment of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation, Tokyo, Japan
| | - Yoshinori Murakami
- Dapartment of Molecular Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Yoichiro Kamatani
- Laboratory of Complex Trail Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukihide Momozawa
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| |
Collapse
|
22
|
Hadi AF, Arta RK, Kushima I, Egawa J, Watanabe Y, Ozaki N, Someya T. Association Analysis of Rare CNTN5 Variants With Autism Spectrum Disorder in a Japanese Population. Neuropsychopharmacol Rep 2025; 45:e12527. [PMID: 39887962 PMCID: PMC11781355 DOI: 10.1002/npr2.12527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Contactin-5 (CNTN5), a neural adhesion molecule involved in synaptogenesis and synaptic maturation in the auditory pathway, has been associated with the pathophysiology of autism spectrum disorder (ASD), particularly hyperacusis. To investigate the role of rare CNTN5 variants in ASD susceptibility, we performed resequencing and association analysis in a Japanese population. METHODS We resequenced the CNTN5 coding regions in 302 patients with ASD and prioritized rare putatively damaging variants. The prioritized variants were then genotyped in 313 patients with ASD and 1065 controls. Subsequently, we conducted an association study of selected variants with ASD in 614 patients with ASD and 61 057 controls. Clinical data were reviewed for patients carrying prioritized variants. RESULTS Through resequencing, we prioritized three rare putatively damaging missense variants (W69G, I227L, and L1000S) in patients with ASD. Although we found a nominally significant association between the I227L variant and ASD, it did not remain significant after post hoc correction. Hyperacusis was found in three out of nine patients carrying prioritized variants. CONCLUSION This study does not provide evidence for the contribution of rare CNTN5 variants to the genetic etiology of ASD in the Japanese population.
Collapse
Affiliation(s)
- Abdul Fuad Hadi
- Department of Psychiatry, School of Medicine, and Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Reza K. Arta
- Department of Psychiatry, School of Medicine, and Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Itaru Kushima
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaAichiJapan
- Medical Genomics CenterNagoya University HospitalNagoyaAichiJapan
| | - Jun Egawa
- Department of Psychiatry, School of Medicine, and Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Yuichiro Watanabe
- Department of Psychiatry, School of Medicine, and Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
- Department of PsychiatryUonuma Kikan HospitalNiigataJapan
| | - Norio Ozaki
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaAichiJapan
| | - Toshiyuki Someya
- Department of Psychiatry, School of Medicine, and Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| |
Collapse
|
23
|
Harris L, McDonagh EM, Zhang X, Fawcett K, Foreman A, Daneck P, Sergouniotis PI, Parkinson H, Mazzarotto F, Inouye M, Hollox EJ, Birney E, Fitzgerald T. Genome-wide association testing beyond SNPs. Nat Rev Genet 2025; 26:156-170. [PMID: 39375560 DOI: 10.1038/s41576-024-00778-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 10/09/2024]
Abstract
Decades of genetic association testing in human cohorts have provided important insights into the genetic architecture and biological underpinnings of complex traits and diseases. However, for certain traits, genome-wide association studies (GWAS) for common SNPs are approaching signal saturation, which underscores the need to explore other types of genetic variation to understand the genetic basis of traits and diseases. Copy number variation (CNV) is an important source of heritability that is well known to functionally affect human traits. Recent technological and computational advances enable the large-scale, genome-wide evaluation of CNVs, with implications for downstream applications such as polygenic risk scoring and drug target identification. Here, we review the current state of CNV-GWAS, discuss current limitations in resource infrastructure that need to be overcome to enable the wider uptake of CNV-GWAS results, highlight emerging opportunities and suggest guidelines and standards for future GWAS for genetic variation beyond SNPs at scale.
Collapse
Affiliation(s)
- Laura Harris
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Ellen M McDonagh
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Xiaolei Zhang
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Katherine Fawcett
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Amy Foreman
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Petr Daneck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Panagiotis I Sergouniotis
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Helen Parkinson
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Francesco Mazzarotto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Michael Inouye
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Edward J Hollox
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Ewan Birney
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Tomas Fitzgerald
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK.
| |
Collapse
|
24
|
Furukawa S, Kushima I, Kato H, Kimura H, Nawa Y, Aleksic B, Banno M, Yamamoto M, Uematsu M, Nagasaki Y, Ogi T, Ozaki N, Ikeda M. Whole-genome sequencing analysis of Japanese autism spectrum disorder trios. Psychiatry Clin Neurosci 2025; 79:87-97. [PMID: 39610113 PMCID: PMC11874045 DOI: 10.1111/pcn.13767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024]
Abstract
AIM Autism spectrum disorder (ASD) is a genetically and phenotypically heterogeneous neurodevelopmental disorder with a strong genetic basis. Conducting the first comprehensive whole-genome sequencing (WGS) analysis of Japanese ASD trios, this study aimed to elucidate the clinical significance of pathogenic variants and enhance the understanding of ASD pathogenesis. METHODS WGS was performed on 57 Japanese patients with ASD and their parents, investigating variants ranging from single-nucleotide variants to structural variants (SVs), short tandem repeats (STRs), mitochondrial variants, and polygenic risk score (PRS). RESULTS Potentially pathogenic variants that could explain observed phenotypes were identified in 18 patients (31.6%) overall and in 10 of 23 patients (43.5%) with comorbid intellectual developmental disorder (IDD). De novo variants in PTEN, CHD7, and HNRNPH2 were identified in patients referred for genetic counseling who exhibited previously reported phenotypes, including one patient with ASD who had profound IDD and macrocephaly with PTEN L320S. Analysis of the AlphaFold3 protein structure indicated potential inhibition of intramolecular interactions within PTEN. SV analysis identified deletions in ARHGAP11B and TMLHE. A pathogenic de novo mitochondrial variant was identified in a patient with ASD who had a history of encephalitis and cognitive decline. GO enrichment analysis of genes with nonsense variants and missense variants (Missense badness, PolyPhen-2, and Constraint >1) showed associations with regulation of growth and ATP-dependent chromatin remodeler activity. No reportable results were obtained in the analysis of STR and PRS. CONCLUSION Characterizing the comprehensive genetic architecture and phenotypes of ASD is a fundamental step towards unraveling its complex biology.
Collapse
Affiliation(s)
- Sawako Furukawa
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Itaru Kushima
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Medical Genomics CenterNagoya University HospitalNagoyaJapan
| | - Hidekazu Kato
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Department of Psychiatry for Parents and ChildrenNagoya University HospitalNagoyaJapan
| | - Hiroki Kimura
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Yoshihiro Nawa
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Department of Psychiatry for Parents and ChildrenNagoya University HospitalNagoyaJapan
| | - Branko Aleksic
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | | | - Maeri Yamamoto
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Mariko Uematsu
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Yukako Nagasaki
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Tomoo Ogi
- Department of Genetics, Research Institute of Environmental Medicine (RIeM)Nagoya UniversityNagoyaJapan
| | - Norio Ozaki
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Pathophysiology of Mental DisordersNagoya University Graduate School of Medicine
| | - Masashi Ikeda
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
25
|
Chen S, Shcherbina A, Schafer ST, Mattingly ZA, Ramesh J, Narayanan C, Banerjee S, Heath B, Regester M, Chen I, Thakurela S, Hallmayer J, O'Hara R, Solomon M, Nordahl CW, Amaral DG, Chetty S. Cellular mechanisms of early brain overgrowth in autistic children: elevated levels of GPX4 and resistance to ferroptosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635706. [PMID: 39975145 PMCID: PMC11838294 DOI: 10.1101/2025.01.30.635706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Autistic individuals with disproportionate megalencephaly (ASD-DM), characterized by enlarged brains relative to body height, have higher rates of intellectual disability and face more severe cognitive challenges than autistic children with average brain sizes. The cellular and molecular mechanisms underlying this neurophenotype remain poorly understood. To investigate these mechanisms, we generated human induced pluripotent stem cells from non-autistic typically developing children and autistic children with and without disproportionate megalencephaly. We assessed these children longitudinally from ages two to twelve years using magnetic resonance imaging and comprehensive cognitive and medical evaluations. We show that neural progenitor cells (NPCs) derived from ASD-DM children exhibit increased rates of cell survival and suppressed cell death, accompanied by heightened oxidative stress and ferrous iron accumulation. Despite these stressors, ASD-DM NPCs actively suppress apoptosis and ferroptosis by regulating proteins such as caspase-3 (CASP3), poly(ADP-ribose) polymerase 1 (PARP1), and glutathione peroxidase 4 (GPX4). Cellular ferroptotic signatures are further supported by elevated expression of selenocysteine genes, including GPX4 , in the blood of ASD-DM children and their mothers, suggesting potential hereditary or environmental influences. Furthermore, we show that peripheral expression of GPX4 and other selenocysteine genes correlate with cognitive outcomes (IQ). These findings underscore the role of ferroptosis in autism, pointing to potential diagnostic biomarkers and targets for intervention.
Collapse
|
26
|
Yoon S, Penzes P. Roles of ANK2/ankyrin-B in neurodevelopmental disorders: Isoform functions and implications for autism spectrum disorder and epilepsy. Curr Opin Neurobiol 2025; 90:102938. [PMID: 39631164 PMCID: PMC11839328 DOI: 10.1016/j.conb.2024.102938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
The ANK2 gene, encoding ankyrin-B, is a high-confidence risk factor for neurodevelopmental disorders (NDDs). Evidence from exome sequencing studies have repeatedly implicated rare variants in ANK2 in autism spectrum disorder. Recently, the functions of ankyrin-B isoforms on neuronal phenotypes have been investigated using a number of techniques including electrophysiology, proteomic screens and behavioral analysis using animal models with loss of distinct Ank2 isoforms or with targeted loss of Ank2 in different cell types and time points during brain development. ANK2 variants and their pathophysiology could provide valuable insights into the molecular mechanisms underlying NDDs. In this review, we focus on recently reported studies to help understand the pathological mechanisms of ANK2 loss and how it may facilitate the development of treatments for NDDs in the future.
Collapse
Affiliation(s)
- Sehyoun Yoon
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Peter Penzes
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Northwestern University, Center for Autism and Neurodevelopment, Chicago, IL, 60611, USA
| |
Collapse
|
27
|
Bulduk BK, Tortajada J, Torres‐Egurrola L, Valiente‐Pallejà A, Martínez‐Leal R, Vilella E, Torrell H, Muntané G, Martorell L. High frequency of mitochondrial DNA rearrangements in the peripheral blood of adults with intellectual disability. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2025; 69:137-152. [PMID: 39506491 PMCID: PMC11735882 DOI: 10.1111/jir.13197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Mitochondrial DNA (mtDNA) rearrangements are recognised factors in mitochondrial disorders and ageing, but their involvement in neurodevelopmental disorders, particularly intellectual disability (ID) and autism spectrum disorder (ASD), remains poorly understood. Previous studies have reported mitochondrial dysfunction in individuals with both ID and ASD. The aim of this study was to investigate the prevalence of large-scale mtDNA rearrangements in ID and ID with comorbid ASD (ID-ASD). METHOD We used mtDNA-targeted next-generation sequencing and the MitoSAlt high-throughput computational pipeline in peripheral blood samples from 76 patients with ID (mean age 52.5 years, 37% female), 59 patients with ID-ASD (mean age 41.3 years, 46% female) and 32 healthy controls (mean age 42.4 years, 47% female) from Catalonia. RESULTS The study revealed a high frequency of mtDNA rearrangements in patients with ID, with 10/76 (13.2%) affected individuals. However, the prevalence was significantly lower in patients with ID-ASD 1/59 (1.7%) and in HC 1/32 (3.1%). Among the mtDNA rearrangements, six were identified as deletions (median size 6937 bp and median heteroplasmy level 2.3%) and six as duplications (median size 10 455 bp and median heteroplasmy level 1.9%). One of the duplications, MT-ATP6 m.8765-8793dup (29 bp), was present in four individuals with ID with a median heteroplasmy level of 3.9%. CONCLUSIONS Our results show that mtDNA rearrangements are frequent in patients with ID, but not in those with ID-ASD, when compared to HC. Additionally, MitoSAlt has demonstrated high sensitivity and accuracy in detecting mtDNA rearrangements, even at very low heteroplasmy levels in blood samples. While the high frequency of mtDNA rearrangements in ID is noteworthy, the role of these rearrangements is currently unclear and needs to be confirmed with further data, particularly in post-mitotic tissues and through age-matched control studies.
Collapse
Affiliation(s)
- B. K. Bulduk
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
| | - J. Tortajada
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
| | - L. Torres‐Egurrola
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
| | - A. Valiente‐Pallejà
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
- CIBER de Salud Mental (CIBERSAM)Instituto de Salud Carlos IIIMadridSpain
| | - R. Martínez‐Leal
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
- CIBER de Salud Mental (CIBERSAM)Instituto de Salud Carlos IIIMadridSpain
- Genètica i Ambient en PsiquiatriaIntellectual Disability and Developmental Disorders Research Unit (UNIVIDD), Fundació VillablancaReusCataloniaSpain
| | - E. Vilella
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
- CIBER de Salud Mental (CIBERSAM)Instituto de Salud Carlos IIIMadridSpain
| | - H. Torrell
- Centre for Omic Sciences (COS)Joint Unit Universitat Rovira i Virgili‐EURECAT Technology Centre of Catalonia, Unique Scientific and Technical InfrastructuresReusCataloniaSpain
| | - G. Muntané
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
- CIBER de Salud Mental (CIBERSAM)Instituto de Salud Carlos IIIMadridSpain
- Institut de Biologia Evolutiva (UPF‐CSIC), Department of Medicine and Life SciencesUniversitat Pompeu Fabra, Parc de Recerca Biomèdica de BarcelonaBarcelonaCataloniaSpain
| | - L. Martorell
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
- CIBER de Salud Mental (CIBERSAM)Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
28
|
Qiu Z, Du A. Revisiting the genetic architecture of autism spectrum disorders in the genomic era: Insights from East Asian studies. Curr Opin Neurobiol 2025; 90:102936. [PMID: 39616786 DOI: 10.1016/j.conb.2024.102936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/28/2024] [Accepted: 11/12/2024] [Indexed: 02/21/2025]
Abstract
This review delves into the genetic landscape of Autism Spectrum Disorder (ASD) in the genomic era, with a special focus on insights from East Asian populations. We analyze a spectrum of genetic research, including whole-exome and whole-genome sequencing, to elucidate both the challenges and advancements in comprehending the genetic foundations of ASD. Critical findings from this review highlight the identification of de novo variants, particularly noting the significant role of rare variants that differ from the common variants identified in earlier research. The review emphasizes the importance of large, diverse, and meticulously maintained ASD cohorts, which are essential for advancing genetic studies and developing potential therapeutic interventions. Through collaborative international efforts, we argue for a global perspective necessary to grasp the intricate genetic factors underlying ASD.
Collapse
Affiliation(s)
- Zilong Qiu
- Department of Neurology, Songjiang Hospital, Songjiang Research Institute, MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ailian Du
- Department of Neurology, Songjiang Hospital, Songjiang Research Institute, MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Winden KD, Ruiz JF, Sahin M. Construction destruction: Contribution of dyregulated proteostasis to neurodevelopmental disorders. Curr Opin Neurobiol 2025; 90:102934. [PMID: 39612590 PMCID: PMC11839335 DOI: 10.1016/j.conb.2024.102934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024]
Abstract
Genetic causes of neurodevelopmental disorders (NDDs) such as epilepsy and autism spectrum disorder are rapidly being uncovered. The genetic risk factors that are responsible for various NDDs fall into many categories, and while some genes such as those involved in synaptic transmission are expected, there are several other classes of genes whose involvement in these disorders is not intuitive. One such group of genes is involved in protein synthesis and degradation, and the balance between these opposing pathways is termed proteostasis. Here, we review these pathways, the genetics of the related neurological disorders, and some potential disease mechanisms. Improved understanding of this collection of genetic disorders will be informative for the pathogenesis of these disorders and imply novel therapeutic strategies.
Collapse
Affiliation(s)
- Kellen D Winden
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Juan F Ruiz
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
30
|
Nishizaki SS, Haghani NK, La GN, Mariano NAF, Uribe-Salazar JM, Kaya G, Regester M, Andrews DS, Nordahl CW, Amaral DG, Dennis MY. m 6A-mRNA Reader YTHDF2 Identified as a Potential Risk Gene in Autism With Disproportionate Megalencephaly. Autism Res 2025. [PMID: 39887636 DOI: 10.1002/aur.3314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 02/01/2025]
Abstract
Among autistic individuals, a subphenotype of disproportionate megalencephaly (ASD-DM) seen at three years of age is associated with co-occurring intellectual disability and poorer prognoses later in life. However, many of the genes contributing to ASD-DM have yet to be delineated. In this study, we identified additional ASD-DM candidate genes with the aim to better define the genetic etiology of this subphenotype of autism. We expanded the previously studied sample size of ASD-DM individuals ten fold by including probands from the Autism Phenome Project and Simons Simplex Collection, totaling 766 autistic individuals meeting the criteria for megalencephaly or macrocephaly and revealing 154 candidate ASD-DM genes harboring de novo protein-impacting variants. Our findings include 14 high confidence autism genes and seven genes previously associated with DM. Five impacted genes have previously been associated with both autism and DM, including CHD8 and PTEN. By performing functional network analysis, we expanded to additional candidate genes, including one previously implicated in ASD-DM (PIK3CA) as well as 184 additional genes connected with ASD or DM alone. Using zebrafish, we modeled a de novo tandem duplication impacting YTHDF2, encoding an N6-methyladenosine (m6A)-mRNA reader, in an ASD-DM proband. Testing zebrafish CRISPR knockdown led to reduced head/brain size, while overexpressing YTHDF2 resulted in increased head/brain size matching that of the proband. Single-cell transcriptomes of YTHDF2 gain-of-function larvae point to reduced expression of Fragile-X-syndrome-associated FMRP-target genes globally and in the developing brain, providing insight into the mechanism underlying autistic phenotypes. We additionally discovered a variant impacting a different gene encoding an m6A reader, YTHDC1, in our ASD-DM cohort. Though we highlight only two cases to date, our study provides support for the m6A-RNA modification pathway as potentially contributing to this severe form of autism.
Collapse
Affiliation(s)
- Sierra S Nishizaki
- Genome Center, University of California, Davis, CA, USA
- Autism Research Training Program, University of California, Davis, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - Nicholas K Haghani
- Genome Center, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
| | - Gabriana N La
- Genome Center, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
| | - Natasha Ann F Mariano
- Genome Center, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
- Postbaccalaureate Research Education Program, University of California, Davis, California, USA
| | - José M Uribe-Salazar
- Genome Center, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
| | - Gulhan Kaya
- Genome Center, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
| | - Melissa Regester
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - Derek Sayre Andrews
- Autism Research Training Program, University of California, Davis, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - Christine Wu Nordahl
- Autism Research Training Program, University of California, Davis, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - David G Amaral
- Autism Research Training Program, University of California, Davis, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - Megan Y Dennis
- Genome Center, University of California, Davis, CA, USA
- Autism Research Training Program, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
| |
Collapse
|
31
|
Biersack B, Nitzsche B, Höpfner M. Histone deacetylases in the regulation of cell death and survival mechanisms in resistant BRAF-mutant cancers. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:6. [PMID: 39935431 PMCID: PMC11810460 DOI: 10.20517/cdr.2024.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/16/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025]
Abstract
Small-molecule BRAF inhibitors (e.g., vemurafenib and dabrafenib) and MEK (MAPK/ERK) kinases inhibitors (e.g., trametinib) have distinctly improved the survival of patients suffering from BRAF-mutant cancers such as melanomas. However, the emergence of resistance to BRAF and MEK inhibitor-based melanoma therapy, as well as the reduced sensitivity of other BRAF-mutant cancers such as CRC, poses a considerable clinical problem. For instance, the reactivation of MAPK/ERK signaling hampering cell death induction mechanisms was responsible for BRAF inhibitor resistance, which can be correlated with distinct post-translational and epigenetic processes. Histone deacetylases (HDACs) are prominent epigenetic drug targets and some HDAC inhibitors have already been clinically approved for the therapy of various blood cancers. In addition, several HDACs were identified, which also play a crucial role in the drug resistance of BRAF-mutant cancers. Consequently, inhibition of HDACs was described as a promising approach to overcome resistance. This review summarizes the influence of HDACs (Zn2+-dependent HDACs and NAD+-dependent sirtuins) on BRAF-mutant cancers and BRAF inhibitor resistance based on upregulated survival mechanisms and the prevention of tumor cell death. Moreover, it outlines reasonable HDAC-based strategies to circumvent BRAF-associated resistance mechanisms based on downregulated cell death mechanisms.
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry Laboratory, University Bayreuth, Bayreuth 95440, Germany
| | - Bianca Nitzsche
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of the Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin 10117, Germany
| | - Michael Höpfner
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of the Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin 10117, Germany
| |
Collapse
|
32
|
Andersen RE, Talukdar M, Sakamoto T, Song JH, Qian X, Lee S, Delgado RN, Zhao S, Eichfeld G, Harms J, Walsh CA. Autism-Associated Genes and Neighboring lncRNAs Converge on Key Gene Regulatory Networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.634000. [PMID: 39896631 PMCID: PMC11785016 DOI: 10.1101/2025.01.20.634000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The diversity of genes implicated in autism spectrum disorder (ASD) creates challenges for identifying core pathophysiological mechanisms. Aggregation of seven different classes of genetic variants implicated in ASD, in a database we call Consensus-ASD, reveals shared features across distinct types of ASD variants. Functional interrogation of 19 ASD genes and 9 neighboring long non-coding RNAs (lncRNAs) using CRISPR-Cas13 strikingly revealed differential gene expression profiles that were significantly enriched for other ASD genes. Furthermore, construction of a gene regulatory network (GRN) enabled the identification of central regulators that exhibit convergently altered activity upon ASD gene disruption. Thus, this study reveals how perturbing distinct ASD-associated genes can lead to shared, broad dysregulation of GRNs with critical relevance to ASD. This provides a crucial framework for understanding how diverse genes, including lncRNAs, can play convergent roles in key neurodevelopmental processes and ultimately contribute to ASD.
Collapse
Affiliation(s)
- Rebecca E. Andersen
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Allen Discovery Center for Human Brain Evolution, Boston, MA, USA
| | - Maya Talukdar
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT MD/PhD Program, Program in Biomedical Informatics, Boston, MA, USA
| | - Tyler Sakamoto
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Janet H.T. Song
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Allen Discovery Center for Human Brain Evolution, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| | - Xuyu Qian
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Allen Discovery Center for Human Brain Evolution, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| | - Seungil Lee
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Ryan N. Delgado
- Department of Genetics, Blavatnik Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Sijing Zhao
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard BBS PhD Program, Boston, MA, USA
| | - Gwenyth Eichfeld
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Colgate University, Hamilton, NY, USA
| | - Julia Harms
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- University of California Berkeley, Berkeley, CA, USA
| | - Christopher A. Walsh
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Allen Discovery Center for Human Brain Evolution, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| |
Collapse
|
33
|
Morton SU, Costain G, French CE, Wakeling E, Szuto A, Christodoulou J, Cohn R, Darras BT, Wojcik MH, D'Gama AM, Dowling JJ, Lunke S, Muntoni F, Raymond L, Rowitch D, Beggs AH, Stark Z, Agrawal PB. Exome and Genome Sequencing to Diagnose the Genetic Basis of Neonatal Hypotonia: An International Consortium Study. Neurology 2025; 104:e210106. [PMID: 39700446 DOI: 10.1212/wnl.0000000000210106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/02/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Hypotonia is a relatively common finding among infants in the neonatal intensive care unit (NICU). Consideration of genetic testing is recommended early in the care of infants with unexplained hypotonia. We aimed to assess the diagnostic yield and overall impact of exome and genome sequencing (ES and GS). METHODS Consecutive infants with hypotonia were identified from research and clinical databases across 5 teaching hospitals in United States, Canada, United Kingdom, and Australia. Inclusion criteria included NICU admission and genetic evaluation. Infants with a known explanation for hypotonia were excluded. Data regarding infant characteristics, genetic testing, and diagnoses were collected. The primary outcome was identification of a molecular diagnosis. Impact on care was a secondary outcome. The Fisher exact and Wilcoxon rank-sum tests were used for statistical analysis. RESULTS We identified 147 infants with unexplained hypotonia. The median gestational age was 39 weeks (interquartile range [IQR] 36-42 weeks), 77 (52%) were female, and the median age was 8 days at the time of evaluation (IQR 2-19 days). Eighty (54%) had hypotonia as the main clinical feature while 67 (46%) had additional multisystem involvement. Seventy-five (51%) underwent rapid ES, 44 (30%) rapid GS, 2 (1%) both ES and GS, and 26 (18%) were admitted before ES or GS became available. Of the 121 infants who underwent ES and/or GS, 72 (60%) had the primary outcome of a molecular diagnosis. In addition, 2 infants with mitochondrial genome variants were diagnosed by mitochondrial GS after negative ES, and one infant needed targeted testing to identify a short tandem repeat expansion missed by GS. The proportion diagnosed by ES and GS was not different between infants with hypotonia as the primary finding (37/56, 66%) and infants with multisystemic symptoms (35/65, 54%, odds ratio [OR] 1.7, CI 0.8-3.7, p value = 0.20). Testing was more likely to have an impact on care for infants receiving a genetic diagnosis (57/66 vs 14/33, OR 8.4, CI 2.9-26.1, p = 1.0E-05). DISCUSSION Rapid ES and GS provided a molecular diagnosis for most of the infants with unexplained hypotonia who underwent testing. Further studies are needed to assess the generalizability of these findings as increased access to genetic testing becomes available. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that in unexplained neonatal hypotonia, rapid ES or GS adds diagnostic specificity.
Collapse
Affiliation(s)
- Sarah U Morton
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Gregory Costain
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Courtney E French
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Emma Wakeling
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Anna Szuto
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - John Christodoulou
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Ronald Cohn
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Basil T Darras
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Monica H Wojcik
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Alissa M D'Gama
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - James J Dowling
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Sebastian Lunke
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Francesco Muntoni
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Lucy Raymond
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - David Rowitch
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Alan H Beggs
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Zornitza Stark
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| | - Pankaj B Agrawal
- From the Division of Newborn Medicine (S.U.M., M.H.W., A.M.D.G.), Boston Children's Hospital; Department of Pediatrics (S.U.M., M.H.W., A.M.D.G., A.H.B., P.B.A.), Harvard Medical School; The Manton Center for Orphan Disease Research (S.U.M., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital; The Broad Institute of MIT and Harvard (S.U.M., M.H.W., A.H.B., P.B.A.), Cambridge, MA; Division of Clinical and Metabolic Genetics (G.C., R.C.), The Hospital for Sick Children; Program in Genetics and Genome Biology (G.C.,. R.C., J.J.D.), SickKids Research Institute; Department of Paediatrics (G.C., R.C., J.J.D.), Department of Molecular Genetics (G.C., A.S., J.J.D.), University of Toronto, Ontario, Canada; Division of Genetics and Genomics (C.E.F., M.H.W., A.H.B., P.B.A.), Boston Children's Hospital, MA; North East Thames Regional Genetic Service (E.W., F.M.), Great Ormond Street Hospital Trust, London, United Kingdom; Department of Genetic Counselling (A.S.), The Hospital for Sick Children, Toronto, OntarioN, Canada; Murdoch Children's Research Institute and Department of Paediatrics (J.C., S.L., Z.S.), University of Melbourne, Victoria; Discipline of Child and Adolescent Health (J.C.), Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Neurology (B.T.D.), Boston Children's Hospital; Epilepsy Genetics Program (A.M.D.G.), Department of Neurology, Boston Children's Hospital, MA; Division of Neurology (J.J.D.), The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pathology (S.L.), University of Melbourne, Australia; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre (F.M.), Great Ormond Street Institute of Child Health, University College London; Departments of Medical Genetics and Paediatrics (L.R., D.R.), University of Cambridge, United Kingdom; Division of Neonatology (D.R.), Department of Pediatrics, UCSF, San Francisco, CA; Australian Genomics Health Alliance (Z.S.); and Division of Neonatology (P.B.A.), Department of Pediatrics, University of Miami and Holtz Children's Hospital, Jackson Health System, FL
| |
Collapse
|
34
|
Avila MN, Jung S, Satterstrom FK, Fu JM, Levy T, Sloofman LG, Klei L, Pichardo T, Stevens CR, Cusick CM, Ames JL, Campos GS, Cerros H, Chaskel R, Costa CIS, Cuccaro ML, Del Pilar Lopez A, Fernandez M, Ferro E, Galeano L, Girardi ACDES, Griswold AJ, Hernandez LC, Lourenço N, Ludena Y, Nuñez DL, Oyama R, Peña KP, Pessah I, Schmidt R, Sweeney HM, Tolentino L, Wang JYT, Albores-Gallo L, Croen LA, Cruz-Fuentes CS, Hertz-Picciotto I, Kolevzon A, Lattig MC, Mayo L, Passos-Bueno MR, Pericak-Vance MA, Siper PM, Tassone F, Trelles MP, Talkowski ME, Daly MJ, Mahjani B, De Rubeis S, Cook EH, Roeder K, Betancur C, Devlin B, Buxbaum JD. Deleterious coding variation associated with autism is consistent across populations, as exemplified by admixed Latin American populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.12.27.24319460. [PMID: 39830258 PMCID: PMC11741445 DOI: 10.1101/2024.12.27.24319460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The past decade has seen remarkable progress in identifying genes that, when impacted by deleterious coding variation, confer high risk for autism spectrum disorder (ASD), intellectual disability, and other developmental disorders. However, most underlying gene discovery efforts have focused on individuals of European ancestry, limiting insights into genetic risks across diverse populations. To help address this, the Genomics of Autism in Latin American Ancestries Consortium (GALA) was formed, presenting here the largest sequencing study of ASD in Latin American individuals (n>15,000). We identified 35 genome-wide significant (FDR < 0.05) ASD risk genes, with substantial overlap with findings from European cohorts, and highly constrained genes showing consistent signal across populations. The results provide support for emerging (e.g., MARK2, YWHAG, PACS1, RERE, SPEN, GSE1, GLS, TNPO3, ANKRD17) and established ASD genes, and for the utility of genetic testing approaches for deleterious variants in diverse populations, while also demonstrating the ongoing need for more inclusive genetic research and testing. We conclude that the biology of ASD is universal and not impacted to any detectable degree by ancestry.
Collapse
Affiliation(s)
- Marina Natividad Avila
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Seulgi Jung
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - F Kyle Satterstrom
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jack M Fu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tess Levy
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Laura G Sloofman
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lambertus Klei
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Thariana Pichardo
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christine R Stevens
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Caroline M Cusick
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jennifer L Ames
- Division of Research, Kaiser Permanente Northern, Pleasanton, California, USA
| | - Gabriele S Campos
- Centro de Estudos do Genoma Humano e Celulas-Tronco, Departamento de Genetica e Biologia Evolutiva, Biociência, Universidade de São Paulo, São Paulo, Brasil
| | - Hilda Cerros
- Division of Research, Kaiser Permanente Northern, Pleasanton, California, USA
| | - Roberto Chaskel
- Facultad de Medicina, Universidad de los Andes, Bogota, Colombia
- Instituto Colombiano del Sistema Nervioso, Clinica Montserrat, Bogota, Colombia
| | - Claudia I S Costa
- Centro de Estudos do Genoma Humano e Celulas-Tronco, Departamento de Genetica e Biologia Evolutiva, Biociência, Universidade de São Paulo, São Paulo, Brasil
| | - Michael L Cuccaro
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Magdalena Fernandez
- Instituto Colombiano del Sistema Nervioso, Clinica Montserrat, Bogota, Colombia
| | - Eugenio Ferro
- Instituto Colombiano del Sistema Nervioso, Clinica Montserrat, Bogota, Colombia
| | - Liliana Galeano
- Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Ana Cristina D E S Girardi
- Centro de Estudos do Genoma Humano e Celulas-Tronco, Departamento de Genetica e Biologia Evolutiva, Biociência, Universidade de São Paulo, São Paulo, Brasil
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Luis C Hernandez
- Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Naila Lourenço
- Centro de Estudos do Genoma Humano e Celulas-Tronco, Departamento de Genetica e Biologia Evolutiva, Biociência, Universidade de São Paulo, São Paulo, Brasil
| | - Yunin Ludena
- MIND (Medical Investigation of Neurodevelopmental Disorders) Institute, University of California Davis, Davis, California, USA
| | - Diana L Nuñez
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- National Center of Posttraumatic Stress Disorders, VA CT Healthcare Center, West Haven, Connecticut, USA
| | - Rosa Oyama
- Centro Ann Sullivan del Peru, Lima, Peru
| | - Katherine P Peña
- Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Isaac Pessah
- MIND (Medical Investigation of Neurodevelopmental Disorders) Institute, University of California Davis, Davis, California, USA
| | - Rebecca Schmidt
- MIND (Medical Investigation of Neurodevelopmental Disorders) Institute, University of California Davis, Davis, California, USA
| | | | | | - Jaqueline Y T Wang
- Centro de Estudos do Genoma Humano e Celulas-Tronco, Departamento de Genetica e Biologia Evolutiva, Biociência, Universidade de São Paulo, São Paulo, Brasil
| | - Lilia Albores-Gallo
- Hospital Psiquiátrico Infantil Dr. Juan N. Navarro, Ciudad de México, Mexico
- Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Lisa A Croen
- Division of Research, Kaiser Permanente Northern, Pleasanton, California, USA
- Kaiser Permanente School of Medicine, Pasadena, California, USA
| | - Carlos S Cruz-Fuentes
- Departamento de Genética, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz México, Ciudad de México, Mexico
| | - Irva Hertz-Picciotto
- MIND (Medical Investigation of Neurodevelopmental Disorders) Institute, University of California Davis, Davis, California, USA
| | - Alexander Kolevzon
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Maria C Lattig
- Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | | | - Maria Rita Passos-Bueno
- Centro de Estudos do Genoma Humano e Celulas-Tronco, Departamento de Genetica e Biologia Evolutiva, Biociência, Universidade de São Paulo, São Paulo, Brasil
| | - Margaret A Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Paige M Siper
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Flora Tassone
- MIND (Medical Investigation of Neurodevelopmental Disorders) Institute, University of California Davis, Davis, California, USA
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Davis, California, USA
| | - M Pilar Trelles
- Psychiatry and Behavioral Sciences, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Michael E Talkowski
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Program in Bioinformatics and Integrative Genomics, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark J Daly
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Behrang Mahjani
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Edwin H Cook
- Department of Psychiatry, University of Illinois Chicago, Chicago, Illinois, USA
| | - Kathryn Roeder
- Department of Statistics, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Catalina Betancur
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, Paris, France
| | - Bernie Devlin
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
35
|
Scala M, Bradley CA, Howe JL, Trost B, Salazar NB, Shum C, Mendes M, Reuter MS, Anagnostou E, MacDonald JR, Ko SY, Frankland PW, Charlebois J, Elsabbagh M, Granger L, Anadiotis G, Pullano V, Brusco A, Keller R, Parisotto S, Pedro HF, Lusk L, McDonnell PP, Helbig I, Mullegama SV, Douine ED, Corona RI, Russell BE, Nelson SF, Graziano C, Schwab M, Simone L, Zara F, Scherer SW. Genetic variants in DDX53 contribute to autism spectrum disorder associated with the Xp22.11 locus. Am J Hum Genet 2025; 112:154-167. [PMID: 39706195 PMCID: PMC11739878 DOI: 10.1016/j.ajhg.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 12/23/2024] Open
Abstract
Autism spectrum disorder (ASD) exhibits an ∼4:1 male-to-female sex bias and is characterized by early-onset impairment of social/communication skills, restricted interests, and stereotyped behaviors. Disruption of the Xp22.11 locus has been associated with ASD in males. This locus includes the three-exon PTCHD1, an adjacent multi-isoform long noncoding RNA (lncRNA) named PTCHD1-AS (spanning ∼1 Mb), and a poorly characterized single-exon RNA helicase named DDX53 that is intronic to PTCHD1-AS. While the relationship between PTCHD1/PTCHD1-AS and ASD is being studied, the role of DDX53 has not been comprehensively examined, in part because there is no apparent functional murine ortholog. Through clinical testing, here, we identified 8 males and 2 females with ASD from 8 unrelated families carrying rare, predicted damaging or loss-of-function variants in DDX53. Additionally, we identified a family consisting of a male proband and his affected mother with high-functioning autism, both harboring a gene deletion involving DDX53 and exons of the noncoding RNA PTCHD1-AS. Then, we examined databases, including the Autism Speaks MSSNG and Simons Foundation Autism Research Initiative, as well as population controls. We identified 26 additional individuals with ASD harboring 19 mostly maternally inherited, rare, damaging DDX53 variations, including two variants detected in families from the original clinical analysis. Our findings in humans support a direct link between DDX53 and ASD, which will be important in clinical genetic testing. These same autism-related findings, coupled with the observation that a functional orthologous gene is not found in mice, may also influence the design and interpretation of murine modeling of ASD.
Collapse
Affiliation(s)
- Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, and Maternal and Child Health, University of Genoa, Genoa, Italy; UOC Genetica Medica, IRCCS Giannina Gaslini, Genoa, Italy.
| | - Clarrisa A Bradley
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | - Jennifer L Howe
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Brett Trost
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Nelson Bautista Salazar
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Carole Shum
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Marla Mendes
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Miriam S Reuter
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Evdokia Anagnostou
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Program in Neurosciences and Mental Health, The Hospital for Sick Children and Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Jeffrey R MacDonald
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Sangyoon Y Ko
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | - Paul W Frankland
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada; Department of Psychology and Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Jessica Charlebois
- Azrieli Centre for Autism Research, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Mayada Elsabbagh
- Azrieli Centre for Autism Research, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Leslie Granger
- Department of Genetics and Metabolism, Randall Children's Hospital, Portland, OR 97227, USA
| | - George Anadiotis
- Department of Genetics and Metabolism, Randall Children's Hospital, Portland, OR 97227, USA
| | - Verdiana Pullano
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Alfredo Brusco
- Department of Neurosciences Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, Italy
| | - Roberto Keller
- Adult Autism Centre DSM ASL Città di Torino, 10138 Turin, Italy
| | - Sarah Parisotto
- Center for Genetic and Genomic Medicine, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Helio F Pedro
- Center for Genetic and Genomic Medicine, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Laina Lusk
- Epilepsy Neurogenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Pamela Pojomovsky McDonnell
- Epilepsy Neurogenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ingo Helbig
- Epilepsy Neurogenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Emilie D Douine
- Department of Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA
| | - Rosario Ivetth Corona
- Department of Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA
| | - Bianca E Russell
- Department of Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA
| | - Stanley F Nelson
- Department of Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA
| | - Claudio Graziano
- Dipartimento MeLabeT, AUSL della Romagna, Pievesestina di Cesena, Cesena, Italy
| | - Maria Schwab
- Center for Genetic and Genomic Medicine, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Laurie Simone
- Center for Genetic and Genomic Medicine, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Federico Zara
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, and Maternal and Child Health, University of Genoa, Genoa, Italy; UOC Genetica Medica, IRCCS Giannina Gaslini, Genoa, Italy
| | - Stephen W Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; McLaughlin Centre, Toronto, ON M5G 0A4, Canada.
| |
Collapse
|
36
|
Mendes M, Chen DZ, Engchuan W, Leal TP, Thiruvahindrapuram B, Trost B, Howe JL, Pellecchia G, Nalpathamkalam T, Alexandrova R, Salazar NB, McKee EA, Rivera-Alfaro N, Lai MC, Bandres-Ciga S, Roshandel D, Bradley CA, Anagnostou E, Sun L, Scherer SW. Chromosome X-wide common variant association study in autism spectrum disorder. Am J Hum Genet 2025; 112:135-153. [PMID: 39706197 PMCID: PMC11739886 DOI: 10.1016/j.ajhg.2024.11.008] [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: 08/05/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024] Open
Abstract
Autism spectrum disorder (ASD) displays a notable male bias in prevalence. Research into rare (<0.1) genetic variants on the X chromosome has implicated over 20 genes in ASD pathogenesis, such as MECP2, DDX3X, and DMD. The "female protective effect" in ASD suggests that females may require a higher genetic burden to manifest symptoms similar to those in males, yet the mechanisms remain unclear. Despite technological advances in genomics, the complexity of the biological nature of sex chromosomes leaves them underrepresented in genome-wide studies. Here, we conducted an X-chromosome-wide association study (XWAS) using whole-genome sequencing data from 6,873 individuals with ASD (82% males) across Autism Speaks MSSNG, Simons Simplex Collection (SSC), and Simons Powering Autism Research (SPARK), alongside 8,981 population controls (43% males). We analyzed 418,652 X chromosome variants, identifying 59 associated with ASD (p values 7.9 × 10-6 to 1.51 × 10-5), surpassing Bonferroni-corrected thresholds. Key findings include significant regions on Xp22.2 (lead SNP rs12687599, p = 3.57 × 10-7) harboring ASB9/ASB11 and another encompassing DDX53 and the PTCHD1-AS long non-coding RNA (lead SNP rs5926125, p = 9.47 × 10-6). When mapping genes within 10 kb of the 59 most significantly associated SNPs, 91 genes were found, 17 of which yielded association with ASD (GRPR, AP1S2, DDX53, HDAC8, PCDH19, PTCHD1, PCDH11X, PTCHD1-AS, DMD, SYAP1, CNKSR2, GLRA2, OFD1, CDKL5, GPRASP2, NXF5, and SH3KBP1). FGF13 emerged as an X-linked ASD candidate gene, highlighted by sex-specific differences in minor allele frequencies. These results reveal significant insights into X chromosome biology in ASD, confirming and nominating genes and pathways for further investigation.
Collapse
Affiliation(s)
- Marla Mendes
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.
| | - Desmond Zeya Chen
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5S 3E3, Canada
| | - Worrawat Engchuan
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Thiago Peixoto Leal
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Bhooma Thiruvahindrapuram
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Brett Trost
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jennifer L Howe
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Giovanna Pellecchia
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Thomas Nalpathamkalam
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Roumiana Alexandrova
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Nelson Bautista Salazar
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Ethan A McKee
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Natalia Rivera-Alfaro
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Meng-Chuan Lai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5G 2C1, Canada; Department of Psychiatry, The Hospital for Sick Children, Toronto, ON M5G 1E8, Canada; Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
| | - Delnaz Roshandel
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Clarrisa A Bradley
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Evdokia Anagnostou
- Autism Research Centre, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON M4G 1R8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lei Sun
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5S 3E3, Canada; Department of Statistical Sciences, Faculty of Arts and Science, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
37
|
Vorstman J, Sebat J, Bourque VR, Jacquemont S. Integrative genetic analysis: cornerstone of precision psychiatry. Mol Psychiatry 2025; 30:229-236. [PMID: 39215185 DOI: 10.1038/s41380-024-02706-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
The role of genetic testing in the domain of neurodevelopmental and psychiatric disorders (NPDs) is gradually changing from providing etiological explanation for the presence of NPD phenotypes to also identifying young individuals at high risk of developing NPDs before their clinical manifestation. In clinical practice, the latter implies a shift towards the availability of individual genetic information predicting a certain liability to develop an NPD (e.g., autism, intellectual disability, psychosis etc.). The shift from mostly a posteriori explanation to increasingly a priori risk prediction is the by-product of the systematic implementation of whole exome or genome sequencing as part of routine diagnostic work-ups during the neonatal and prenatal periods. This rapid uptake of genetic testing early in development has far-reaching consequences for psychiatry: Whereas until recently individuals would come to medical attention because of signs of abnormal developmental and/or behavioral symptoms, increasingly, individuals are presented based on genetic liability for NPD outcomes before NPD symptoms emerge. This novel clinical scenario, while challenging, also creates opportunities for research on prevention interventions and precision medicine approaches. Here, we review why optimization of individual risk prediction is a key prerequisite for precision medicine in the sphere of NPDs, as well as the technological and statistical methods required to achieve this ambition.
Collapse
Affiliation(s)
- Jacob Vorstman
- Department of Psychiatry, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada.
| | - Jonathan Sebat
- Department of Psychiatry, Department of Cellular & Molecular Medicine, Beyster Center of Psychiatric Genomics, University of California San Diego, San Diego, CA, USA
| | - Vincent-Raphaël Bourque
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Sébastien Jacquemont
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC, Canada
- Département de Pédiatrie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
38
|
Wong NR, Klomhaus A, Adams DJ, Schneider BN, Mehta S, DiStefano C, Wilson RB, Martinez-Agosto JA, Jeste SS, Besterman AD. Clinical factors associated with genetic diagnosis in suspected neurogenetic disorders in a tertiary care clinic. Genet Med 2025; 27:101252. [PMID: 39395029 PMCID: PMC11717587 DOI: 10.1016/j.gim.2024.101252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 10/14/2024] Open
Abstract
PURPOSE This study aimed to identify phenotypic factors associated with genetic diagnoses in patients with neurodevelopmental disorders and generate a decision tree to assist clinicians in identifying patients most likely to receive a positive result on genetic testing. METHODS We retrospectively reviewed the charts of 316 patients evaluated in a neurodevelopmental clinic between 2014 and 2019. Patients were categorized based on genetic test results. Analyses were performed to identify variables that discriminate between patients with and without a genetic diagnosis. RESULTS Patients with a genetic diagnosis were more likely to be female and have a history of motor delay, hypotonia, congenital heart disease, and early intervention. Classification and regression tree analysis revealed that 75% of patients with motor delay had a genetic diagnosis. In patients without motor delay, hypotonia, age of walking, and age at initial evaluation were important indicators of a genetic diagnosis. CONCLUSION Our findings suggest that motor delay and hypotonia are associated with genetic diagnoses in children with neurodevelopmental disorders. The decision tree highlights patient subsets at greater risk and suggests possible phenotypic screens. Future studies could develop validated decision trees based on phenotypic data to assist clinicians in stratifying patients for genetic testing.
Collapse
Affiliation(s)
- Nicole R Wong
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Alexandra Klomhaus
- Department of General Internal Medicine and Health Services Research, University of California, Los Angeles, CA
| | - David J Adams
- University of California Los Angeles, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA; University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | - Benjamin N Schneider
- University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA; University of California Los Angeles, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Los Angeles, CA
| | | | - Charlotte DiStefano
- Children's Hospital Los Angeles, Department of Psychiatry, Los Angeles, CA; University of Southern California, Department of Psychiatry and The Biobehavioral Sciences, Los Angeles, CA
| | - Rujuta B Wilson
- University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA; University of California Los Angeles, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Los Angeles, CA
| | - Julian A Martinez-Agosto
- University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA; University of California Los Angeles, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Los Angeles, CA; University of California Los Angeles, Department of Human Genetics, Los Angeles, CA
| | - Shafali S Jeste
- Children's Hospital Los Angeles, Division of Neurology, Los Angeles, CA; University of Southern California, Departments of Pediatrics and Neurology, Los Angeles, CA
| | - Aaron D Besterman
- University of California San Diego Department of Psychiatry, Division of Child and Adolescent Psychiatry, San Diego, CA; Rady Children's Hospital of San Diego, San Diego, CA; Rady Children's Institute for Genomic Medicine, San Diego, CA.
| |
Collapse
|
39
|
Gupta V, Ben-Mahmoud A, Idris AB, Hottenga JJ, Habbab W, Alsayegh A, Kim HG, AL-Mamari W, Stanton LW. Genetic Variant Analyses Identify Novel Candidate Autism Risk Genes from a Highly Consanguineous Cohort of 104 Families from Oman. Int J Mol Sci 2024; 25:13700. [PMID: 39769462 PMCID: PMC11679916 DOI: 10.3390/ijms252413700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Deficits in social communication, restricted interests, and repetitive behaviours are hallmarks of autism spectrum disorder (ASD). Despite high genetic heritability, the majority of clinically diagnosed ASD cases have unknown genetic origins. We performed genome sequencing on mothers, fathers, and affected individuals from 104 families with ASD in Oman, a Middle Eastern country underrepresented in international genetic studies. This approach identified 48 novel candidate genes significantly associated with ASD in Oman. In particular, 35 of these genes have been previously implicated in neurodevelopmental disorders (NDDs) in other populations, underscoring the conserved genetic basis of ASD across ethnicities. Genetic variants within these candidate genes that would impact the encoded protein included 1 insertion, 4 frameshift, 6 splicing, 12 nonsense, and 67 missense changes. Notably, 61% of the SNVs were homozygous, suggesting a prominent recessive genetic architecture for ASD in this unique population. The scarcity of genetic studies on ASD in the Arabian Peninsula has impeded the understanding of the unique genetic landscape of ASD in this region. These findings help bridge this knowledge gap and provide valuable insights into the complex genetic basis of ASD in Oman.
Collapse
Affiliation(s)
- Vijay Gupta
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha P.O. Box 5825, Qatar; (V.G.); (A.B.-M.); (J.-J.H.); (W.H.); (H.-G.K.)
| | - Afif Ben-Mahmoud
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha P.O. Box 5825, Qatar; (V.G.); (A.B.-M.); (J.-J.H.); (W.H.); (H.-G.K.)
| | - Ahmed B. Idris
- Developmental Paediatric Unit, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat 123, Oman;
| | - Jouke-Jan Hottenga
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha P.O. Box 5825, Qatar; (V.G.); (A.B.-M.); (J.-J.H.); (W.H.); (H.-G.K.)
- Department of Biological Psychology, Vrije Universiteit Amsterdam, 1081 BT Amsterdam, The Netherlands
| | - Wesal Habbab
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha P.O. Box 5825, Qatar; (V.G.); (A.B.-M.); (J.-J.H.); (W.H.); (H.-G.K.)
| | - Abeer Alsayegh
- Genomics Department, Sultan Qaboos Comprehensive Cancer Care and Research Center, University Medical City, Muscat 123, Oman;
| | - Hyung-Goo Kim
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha P.O. Box 5825, Qatar; (V.G.); (A.B.-M.); (J.-J.H.); (W.H.); (H.-G.K.)
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Watfa AL-Mamari
- Developmental Paediatric Unit, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat 123, Oman;
| | - Lawrence W. Stanton
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha P.O. Box 5825, Qatar; (V.G.); (A.B.-M.); (J.-J.H.); (W.H.); (H.-G.K.)
- College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha P.O. Box 5825, Qatar
| |
Collapse
|
40
|
Danieli PP, Hoang N, Selvanayagam T, Yang A, Breetvelt E, Tabbers M, Cohen C, Aelvoet AS, Trost B, Ward T, Semotiuk K, Durno C, Aronson M, Cohen Z, Dekker E, Vorstman J. Autistic traits in youth with familial adenomatous polyposis: A Dutch-Canadian case-control study. Am J Med Genet B Neuropsychiatr Genet 2024; 195:e32999. [PMID: 38967411 DOI: 10.1002/ajmg.b.32999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 07/06/2024]
Abstract
This study investigated the neurodevelopmental impact of pathogenic adenomatous polyposis coli (APC) gene variants in patients with familial adenomatous polyposis (FAP), a cancer predisposition syndrome. We hypothesized that certain pathogenic APC variants result in behavioral-cognitive challenges. We compared 66 FAP patients (cases) and 34 unaffected siblings (controls) to explore associations between APC variants and behavioral and cognitive challenges. Our findings indicate that FAP patients exhibited higher Social Responsiveness Scale (SRS) scores, suggesting a greater prevalence of autistic traits when compared to unaffected siblings (mean 53.8 vs. 47.4, Wilcoxon p = 0.018). The distribution of SRS scores in cases suggested a bimodal pattern, potentially linked to the location of the APC variant, with scores increasing from the 5' to 3' end of the gene (Pearson's r = 0.33, p = 0.022). While we observed a trend toward lower educational attainment in cases, this difference was not statistically significant. This study is the first to explore the connection between APC variant location and neurodevelopmental traits in FAP, expanding our understanding of the genotype-phenotype correlation. Our results emphasize the importance of clinical assessment for autistic traits in FAP patients, shedding light on the potential role of APC gene variants in these behavioral and cognitive challenges.
Collapse
Affiliation(s)
- Polina Perlman Danieli
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ny Hoang
- Department of Genetic Counselling, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Thanuja Selvanayagam
- Department of Genetic Counselling, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alvin Yang
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Familial Gastrointestinal Cancer Registry at the Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Elemi Breetvelt
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Merit Tabbers
- Department of Pediatrics, Emma Children's Hospital, Amsterdam, The Netherlands
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Emma Children's Hospital, Amsterdam Reproduction and Development and Amsterdam Gastroenterology Endocrinology Metabolism Research Institutes, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Christine Cohen
- Department of Gastroenterology and Hepatology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Arthur S Aelvoet
- Department of Gastroenterology and Hepatology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Brett Trost
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Thomas Ward
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- The Familial Gastrointestinal Cancer Registry at the Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Kara Semotiuk
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- The Familial Gastrointestinal Cancer Registry at the Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Carol Durno
- The Familial Gastrointestinal Cancer Registry at the Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Gastroenterology/Hepatology & Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Melyssa Aronson
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- The Familial Gastrointestinal Cancer Registry at the Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Zane Cohen
- The Familial Gastrointestinal Cancer Registry at the Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Evelien Dekker
- Department of Gastroenterology and Hepatology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jacob Vorstman
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Developmental Psychopathology, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Shum C, Han SY, Thiruvahindrapuram B, Wang Z, de Rijke J, Zhang B, Sundberg M, Chen C, Buttermore ED, Makhortova N, Howe J, Sahin M, Scherer SW. Combining Off-flow, a Nextflow-coded program, and whole genome sequencing reveals unintended genetic variation in CRISPR/Cas-edited iPSCs. Comput Struct Biotechnol J 2024; 23:638-647. [PMID: 38283851 PMCID: PMC10819409 DOI: 10.1016/j.csbj.2023.12.036] [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: 10/31/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/30/2024] Open
Abstract
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas nucleases and human induced pluripotent stem cell (iPSC) technology can reveal deep insight into the genetic and molecular bases of human biology and disease. Undesired editing outcomes, both on-target (at the edited locus) and off-target (at other genomic loci) hinder the application of CRISPR-Cas nucleases. We developed Off-flow, a Nextflow-coded bioinformatic workflow that takes a specific guide sequence and Cas protein input to call four separate off-target prediction programs (CHOPCHOP, Cas-Offinder, CRISPRitz, CRISPR-Offinder) to output a comprehensive list of predicted off-target sites. We applied it to whole genome sequencing (WGS) data to investigate the occurrence of unintended effects in human iPSCs that underwent repair or insertion of disease-related variants by homology-directed repair. Off-flow identified a 3-base-pair-substitution and a mono-allelic genomic deletion at the target loci, KCNQ2, in 2 clones. Unbiased WGS analysis further identified off-target missense variants and a mono-allelic genomic deletion at the targeted locus, GNAQ, in 10 clones. On-target substitution and deletions had escaped standard PCR and Sanger sequencing analysis, while missense variants at other genomic loci were not detected by Off-flow. We used these results to filter out iPSC clones for subsequent functional experiments. Off-flow, which we make publicly available, works for human and mouse genomes currently and can be adapted for other genomes. Off-flow and WGS analysis can improve the integrity of studies using CRISPR/Cas-edited cells and animal models.
Collapse
Affiliation(s)
- Carole Shum
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Sang Yeon Han
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | | | - Zhuozhi Wang
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Jill de Rijke
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Benjamin Zhang
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Maria Sundberg
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cidi Chen
- Human Neuron Core, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Nina Makhortova
- Human Neuron Core, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jennifer Howe
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Mustafa Sahin
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen W. Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics and McLaughlin Centre, University of Toronto, Toronto, ON M5S 1A8, Canada
- Lead contact
| |
Collapse
|
42
|
Rong M, Zulfiqar Ali Q, Aledo-Serrano A, Bayat A, Devinsky O, Qaiser F, Chandran I, Ali A, Fasano A, Bassett AS, Andrade DM. Adult Phenotype of CHD2-Associated Disorders. Neurol Genet 2024; 10:e200194. [PMID: 39601014 PMCID: PMC11595326 DOI: 10.1212/nxg.0000000000200194] [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: 04/10/2024] [Accepted: 08/26/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives Pathogenic CHD2 variants are associated with neurodevelopmental disorders and developmental and epileptic encephalopathy. While pediatric CHD2 phenotypes have been readily explored, adult phenotypes are not well understood. We aimed to investigate the phenotypic spectrum of adult patients with CHD2 variants. Methods Patients 18 years or older with likely pathogenic or pathogenic (LP/P) CHD2 variants were included. We used standardized tools to evaluate current seizures, medication use, sleep, gastrointestinal symptoms, pain response, gait, social communication disorder, and adaptive behavioral skills of patients. Results In this prospective study, 14 unrelated adult patients (age range: 18-45 years, median: 21 years) with LP/P CHD2 variants were described. Eleven novel variants were identified. No genotype-phenotype correlations were identified. 79% of adults still have ongoing seizures. Photosensitivity was present in 64% of adult patients. Autism spectrum disorder was diagnosed in 71% of patients. Only 29% were able to read and understand material at a sixth-grade level or higher. Behavioral issues were reported in 100% of adult patients, and 71% had internalizing features, such as anxiety. Self-injurious behaviors were present in 50%. Only 43% could ambulate independently. Additional characteristics included reflux (36%), constipation (71%), and abnormal pain responsiveness (43%). 1 patient presented with nonepileptic breath-holding spells leading to cyanosis. No patient could perform all basic activities of daily living independently, all the time. Higher seizure severity was associated with worse nonseizure outcomes (p = 0.04). Discussion Most adults with CHD2 continue to have seizures, and seizure severity is associated with worse comorbidities such as maladaptive behaviors, gait, gastrointestinal, sleep, and abnormal pain responsiveness. Longevity has not been systematically studied in this group of patients. Here we describe a group of adult patients (up to 45 years of age) and the natural history of this condition. These data may provide prognostic insights for families of pediatric patients and help identify key points to be addressed in future precision trials for patients with CHD2 variants.
Collapse
Affiliation(s)
- Marlene Rong
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| | - Quratulain Zulfiqar Ali
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| | - Angel Aledo-Serrano
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| | - Allan Bayat
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| | - Orrin Devinsky
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| | - Farah Qaiser
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| | - Ilakkiah Chandran
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| | - Anum Ali
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| | - Alfonso Fasano
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| | - Anne S Bassett
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| | - Danielle M Andrade
- From the Institute of Medical Science (M.R.), University of Toronto; Adult Genetic Epilepsy (AGE) Program (M.R., Q.Z.A., F.Q., I.C., A.A., D.M.A.), Krembil Neurosciences Institute, Toronto Western Hospital, University Health Network, Canada; Epilepsy Unit (A.A.-S.), Vithas Clinical Neuroscience Institute, Vithas Madrid University Hospitals; Faculty of Experimental Sciences (A.A.-S.), Francisco de Vitoria University, Madrid, Spain; Department of Drug Design and Pharmacology (A.B.), University of Copenhagen; Department for Genetics and Personalized Medicine (A.B.), Danish Epilepsy Centre, Dianalund; Institute for Regional Health Services (A.B.), University of Southern Denmark, Odense; NYU Langone Epilepsy Center (O.D., F.Q., A.A.); Edmond J. Safra Program in Parkinson's Disease (A.F.), Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN; Division of Neurology (A.F., D.M.A.), Department of Medicine, University of Toronto; Krembil Brain Institute (A.F., D.M.A.); Clinical Genetics Research Program (A.S.B.), Centre for Addiction and Mental Health; The Dalglish Family 22q Clinic (A.S.B.), Toronto General Hospital, University Health Network; Department of Psychiatry (A.S.B.), University of Toronto, Ontario; Toronto Congenital Cardiac Centre for Adults (A.S.B.), Division of Cardiology, Department of Medicine, and Department of Psychiatry, University Health Network and Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute (A.S.B.), Toronto, Ontario, Canada
| |
Collapse
|
43
|
Zhang F, Liu L, Peng J, Ding G, Li Y, Biswal BB, Wang P. Transdiagnostic and Diagnosis-Specific Morphological Similarity Related Transcriptional Profile in Attention-Deficit/Hyperactivity Disorder and Autism Spectrum Disorder. J Am Acad Child Adolesc Psychiatry 2024:S0890-8567(24)02022-7. [PMID: 39608637 DOI: 10.1016/j.jaac.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 08/27/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVE Attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) are both highly heritable developmental psychiatric disorders and exhibit a high degree of comorbidity. Our objective is to enhance understanding of the transdiagnostic and diagnosis-specific structural alterations and related cellular and genetic pathophysiological mechanisms between ADHD and ASD. METHOD We used structural magnetic resonance imaging data of 247 subjects from the publicly available 1000 Functional Connectomes Project, including 91 individuals with ADHD, 49 individuals with ASD, and 107 age- and sex-matched controls. We performed morphological similarity networks (MSN) and gene transcriptional profile analysis on these image data to identify the anatomical changes and MSN-related genes. Enrichment analysis was further conducted on ADHD/ASD risk genes and MSN-related genes. RESULTS Individuals with ADHD showed the diagnosis-specific MSN changes distributing in areas related to high-level cognitive functions, whereas ASD had MSN changes in areas related to language comprehension and spatial location. ADHD and ASD exhibited the transdiagnostic morphological increase in the right middle temporal gyrus. Gene transcriptional profile analysis showed enrichment of ADHD and ASD risk genes in more than 10 biological processes, primarily including function of synapse transmission and development. Genes in excitatory and inhibitory neurons also enriched in pathways with similar function. CONCLUSION The transdiagnostic morphological dedifferentiation in the right middle temporal gyrus might indicate the shared motion impairments in ADHD and ASD. Evidence from the transcription of MSN-related genes further indicates a potential imbalance in excitatory and inhibitory neural pathways in ADHD and ASD. DIVERSITY & INCLUSION STATEMENT We worked to ensure sex and gender balance in the recruitment of human participants. We worked to ensure race, ethnic, and/or other types of diversity in the recruitment of human participants. We worked to ensure that the study questionnaires were prepared in an inclusive way. We worked to ensure sex balance in the selection of non-human subjects. We worked to ensure diversity in experimental samples through the selection of the cell lines. We worked to ensure diversity in experimental samples through the selection of the genomic datasets. Diverse cell lines and/or genomic datasets were not available. We actively worked to promote sex and gender balance in our author group. We actively worked to promote inclusion of historically underrepresented racial and/or ethnic groups in science in our author group. While citing references scientifically relevant for this work, we also actively worked to promote inclusion of historically underrepresented racial and/or ethnic groups in science in our reference list. While citing references scientifically relevant for this work, we also actively worked to promote sex and gender balance in our reference list.
Collapse
Affiliation(s)
- Fanyu Zhang
- University of Electronic Science and Technology of China, Chengdu, China
| | - Lin Liu
- University of Electronic Science and Technology of China, Chengdu, China
| | - Jinzhong Peng
- University of Electronic Science and Technology of China, Chengdu, China
| | - Guobin Ding
- University of Electronic Science and Technology of China, Chengdu, China
| | - Yilu Li
- University of Electronic Science and Technology of China, Chengdu, China
| | - Bharat B Biswal
- University of Electronic Science and Technology of China, Chengdu, China; New Jersey Institute of Technology, Newark, New Jersey
| | - Pan Wang
- University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
44
|
Eisfeldt J, Higginbotham EJ, Lenner F, Howe J, Fernandez BA, Lindstrand A, Scherer SW, Feuk L. Resolving complex duplication variants in autism spectrum disorder using long-read genome sequencing. Genome Res 2024; 34:1763-1773. [PMID: 39472019 DOI: 10.1101/gr.279263.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/27/2024] [Indexed: 11/22/2024]
Abstract
Rare or de novo structural variation, primarily in the form of copy number variants, is detected in 5%-10% of autism spectrum disorder (ASD) families. While complex structural variants involving duplications can generally be detected using microarray or short-read genome sequencing (GS), these methods frequently fail to characterize breakpoints at nucleotide resolution, requiring additional molecular methods for validation and fine-mapping. Here, we use Oxford Nanopore Technologies PromethION long-read GS to characterize complex genomic rearrangements (CGRs) involving large duplications that segregate with ASD in five families. In total, we investigated 13 CGR carriers and were able to resolve all breakpoint junctions at nucleotide resolution. While all breakpoints were identified, the precise genomic architecture of one rearrangement remained unresolved with three different potential structures. The findings in two families include potential fusion genes formed through duplication rearrangements, involving IL1RAPL1-DMD and SUPT16H-CHD8 In two of the families originating from the same geographical region, an identical rearrangement involving ANK2 was identified, which likely represents a founder variant. In addition, we analyze methylation status directly from the long-read data, allowing us to assess the activity of rearranged genes and regulatory regions. Investigation of methylation across the CGRs reveals aberrant methylation status in carriers across a rearrangement affecting the CREBBP locus. In aggregate, our results demonstrate the utility of nanopore sequencing to pinpoint CGRs associated with ASD in five unrelated families, and highlight the importance of a gene-centric description of disease-associated complex chromosomal rearrangements.
Collapse
Affiliation(s)
- Jesper Eisfeldt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden
- Department of Clinical Genetics and Genomics, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Edward J Higginbotham
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Felix Lenner
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory Uppsala, Uppsala University, 751 85 Uppsala, Sweden
| | - Jennifer Howe
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Bridget A Fernandez
- Department of Pediatrics and The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Los Angeles, California 90033, USA
- Discipline of Genetics, Faculty of Medicine, Memorial University of Newfoundland, St John's, Newfoundland and Labrador A1B 3V6, Canada
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden
- Department of Clinical Genetics and Genomics, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Stephen W Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Molecular Genetics, McLaughlin Centre, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Lars Feuk
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory Uppsala, Uppsala University, 751 85 Uppsala, Sweden;
| |
Collapse
|
45
|
Chang S, Liu JJ, Zhao Y, Pang T, Zheng X, Song Z, Zhang A, Gao X, Luo L, Guo Y, Liu J, Yang L, Lu L. Whole-genome sequencing identifies novel genes for autism in Chinese trios. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2368-2381. [PMID: 39126614 DOI: 10.1007/s11427-023-2564-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/16/2024] [Indexed: 08/12/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with high genetic heritability but heterogeneity. Fully understanding its genetics requires whole-genome sequencing (WGS), but the ASD studies utilizing WGS data in Chinese population are limited. In this study, we present a WGS study for 334 individuals, including 112 ASD patients and their non-ASD parents. We identified 146 de novo variants in coding regions in 85 cases and 60 inherited variants in coding regions. By integrating these variants with an association model, we identified 33 potential risk genes (P<0.001) enriched in neuron and regulation related biological process. Besides the well-known ASD genes (SCN2A, NF1, SHANK3, CHD8 etc.), several high confidence genes were highlighted by a series of functional analyses, including CTNND1, DGKZ, LRP1, DDN, ZNF483, NR4A2, SMAD6, INTS1, and MRPL12, with more supported evidence from GO enrichment, expression and network analysis. We also integrated RNA-seq data to analyze the effect of the variants on the gene expression and found 12 genes in the individuals with the related variants had relatively biased expression. We further presented the clinical phenotypes of the proband carrying the risk genes in both our samples and Caucasian samples to show the effect of the risk genes on phenotype. Regarding variants in non-coding regions, a total of 74 de novo variants and 30 inherited variants were predicted as pathogenic with high confidence, which were mapped to specific genes or regulatory features. The number of de novo variants found in patient was significantly associated with the parents' ages at the birth of the child, and gender with trend. We also identified small de novo structural variants in ASD trios. The results in this study provided important evidence for understanding the genetic mechanism of ASD.
Collapse
Affiliation(s)
- Suhua Chang
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
- Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, Beijing, 100191, China
| | - Jia Jia Liu
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
- School of Nursing, Peking University, Beijing, 100191, China
| | - Yilu Zhao
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | - Tao Pang
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | - Xiangyu Zheng
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | | | - Anyi Zhang
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | - Xuping Gao
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | - Lingxue Luo
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | - Yanqing Guo
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China.
| | - Jing Liu
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China.
| | - Li Yang
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China.
| | - Lin Lu
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China.
- Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, Beijing, 100191, China.
- National Institute on Drug Dependence, Peking University, Beijing, 100191, China.
| |
Collapse
|
46
|
Leblond CS, Rolland T, Barthome E, Mougin Z, Fleury M, Ecker C, Bonnot-Briey S, Cliquet F, Tabet AC, Maruani A, Chaumette B, Green J, Delorme R, Bourgeron T. A Genetic Bridge Between Medicine and Neurodiversity for Autism. Annu Rev Genet 2024; 58:487-512. [PMID: 39585908 DOI: 10.1146/annurev-genet-111523-102614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Autism represents a large spectrum of diverse individuals with varying underlying genetic architectures and needs. For some individuals, a single de novo or ultrarare genetic variant has a large effect on the intensity of specific dimensions of the phenotype, while, for others, a combination of thousands of variants commonly found in the general population are involved. The variants with large impact are found in up to 30% of autistic individuals presenting with intellectual disability, significant speech delay, motor delay, and/or seizures. The common variants are shared with those found in individuals with attention-deficit/hyperactivity disorder, major depressive disorders, greater educational attainment, and higher cognitive performance, suggesting overlapping genetic architectures. The genetic variants modulate the function of chromatin remodeling and synaptic proteins that influence the connectivity of neuronal circuits and, in interaction with the environment of each individual, the subsequent cognitive and personal trajectory of the child. Overall, this genetic heterogeneity mirrors the phenotypic diversity of autistic individuals and provides a helpful bridge between biomedical and neurodiversity perspectives. We propose that participative and multidisciplinary research should use this information to understand better the assessment, treatments, and accommodations that individuals with autism and families need.
Collapse
Affiliation(s)
- Claire S Leblond
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
| | - Thomas Rolland
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
| | - Eli Barthome
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
| | - Zakaria Mougin
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
| | - Mathis Fleury
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
| | - Christine Ecker
- Department of Child and Adolescent Psychiatry, University Hospital of the Goethe University, Frankfurt am Main, Germany
| | | | - Freddy Cliquet
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
| | - Anne-Claude Tabet
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
- Department of Genetics, Cytogenetics Unit, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Anna Maruani
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
- Department of Child and Adolescent Psychiatry, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Boris Chaumette
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
- Groupe Hospitalier Universitaire-Paris Psychiatry and Neurosciences, Hôpital Sainte Anne, Paris, France
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Université Paris Cité, Paris, France
| | - Jonathan Green
- Division of Psychology and Mental Health, University of Manchester and Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Richard Delorme
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
- Department of Child and Adolescent Psychiatry, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions, Institut Pasteur, CNRS UMR3571, Institut Universitaire de France, Université Paris Cité, Paris, France;
| |
Collapse
|
47
|
Caiola HO, Wu Q, Li J, Wang XF, Soni S, Monahan K, Wagner GC, Pang ZP, Zhang H. Neuronal connectivity, behavioral, and transcriptional alterations associated with the loss of MARK2. FASEB J 2024; 38:e70124. [PMID: 39436150 DOI: 10.1096/fj.202400454r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/03/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
Neuronal connectivity is essential for adaptive brain responses and can be modulated by dendritic spine plasticity and the intrinsic excitability of individual neurons. Dysregulation of these processes can lead to aberrant neuronal activity, which has been associated with numerous neurological disorders including autism, epilepsy, and Alzheimer's disease. Nonetheless, the molecular mechanisms underlying abnormal neuronal connectivity remain unclear. We previously found that the serine/threonine kinase Microtubule Affinity Regulating Kinase 2 (MARK2), also known as Partitioning Defective 1b (Par1b), is important for the formation of dendritic spines in vitro. However, despite its genetic association with several neurological disorders, the in vivo impact of MARK2 on neuronal connectivity and cognitive functions remains unclear. Here, we demonstrate that the loss of MARK2 in vivo results in changes to dendritic spine morphology, which in turn leads to a decrease in excitatory synaptic transmission. Additionally, the loss of MARK2 produces substantial impairments in learning and memory, reduced anxiety, and defective social behavior. Notably, MARK2 deficiency results in heightened seizure susceptibility. Consistent with this observation, electrophysiological analysis of hippocampal slices indicates underlying neuronal hyperexcitability in MARK2-deficient neurons. Finally, RNAseq analysis reveals transcriptional changes in genes regulating synaptic transmission and ion homeostasis. These results underscore the in vivo role of MARK2 in governing synaptic connectivity, neuronal excitability, and cognitive functions.
Collapse
Affiliation(s)
- Hanna O Caiola
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Qian Wu
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Junlong Li
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Xue-Feng Wang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Shaili Soni
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Kevin Monahan
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, USA
| | - George C Wagner
- Department of Psychology, Rutgers University, Piscataway, New Jersey, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| |
Collapse
|
48
|
Wang L, Yu L, Liu Z, Che C, Wang Y, Zhao Y, Zhu M, Yang G, Cao A. FMT intervention decreases urine 5-HIAA levels: a randomized double-blind controlled study. Front Med (Lausanne) 2024; 11:1411089. [PMID: 39493719 PMCID: PMC11529335 DOI: 10.3389/fmed.2024.1411089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
Background Autism spectrum disorder (ASD) is often linked to gastrointestinal issues and altered serotonin metabolism. Emerging evidence suggests gut microbiota influence both, with fecal microbiota transplantation (FMT) offering a potential therapeutic approach. However, its impact on serotonin metabolism and ASD symptoms is not well understood. In this study, we aimed to evaluate the clinical effects of FMT and examine changes in specific urinary metabolites in children with ASD. Methods A randomized double-blind controlled trial was performed to evaluate the clinical effects of FMT on GI and ASD-related symptoms. Gastrointestinal symptoms were assessed using the Gastrointestinal Symptom Rating Scale (GSRS), and the ASD-related symptoms were assessed using the Childhood Autism Rating Scale (CARS), Aberrant Behavior Checklist (ABC), and Social Responsiveness Scale (SRS) scores. Urinary metabolites were analyzed by homogeneous enzyme immunoassay using commercially available kits. Results Significant improvements in GI and core ASD symptoms were observed following FMT intervention. The average GSRS scores decreased from 30.17 (before) to 19 (after; p < 0.0001), CARS scores decreased from 36.22 to 33.33 (p < 0.0001), SRS scores decreased from 151.17 to 137.5 (p = 0.0002), and the ABC scores decreased 76.39 to 53.17 (p < 0.0001) in the FMT group. However, in the placebo group, GSRS, CARS, and SRS scores showed no significant changes, while ABC scores decreased from 72 to 58.75 (p = 0.034). The FMT group also showed a significant reduction in urinary 5-hydroxyindoleacetic acid (5-HIAA) levels from 8.6 to 7.32 mg/L (p = 0.022), while other metabolites showed no significant changes. Conclusion FMT is a safe and effective treatment for improving GI and core symptoms in children with ASD, with 5-HIAA showing potential as a urinary biomarker for treatment response.
Collapse
Affiliation(s)
- Lihong Wang
- Department of Pediatrics, Shandong University Qilu Hospital, Shandong, Jinan, China
| | - Lianhu Yu
- Department of Pediatrics, Shandong University Qilu Hospital, Shandong, Jinan, China
| | - Zhiyue Liu
- Department of Pediatrics, Shandong University Qilu Hospital, Shandong, Jinan, China
| | - Chao Che
- Department of Pediatrics, Shandong University Qilu Hospital, Shandong, Jinan, China
| | - Yu Wang
- Department of Pediatrics, Shandong University Qilu Hospital, Shandong, Jinan, China
| | - Yongheng Zhao
- Department of Pediatrics, Shandong University Qilu Hospital, Shandong, Jinan, China
| | - Mengna Zhu
- Department of Pediatrics, Shandong University Qilu Hospital, Shandong, Jinan, China
| | - Guang Yang
- Department of Pediatrics, Chinese PLA General Hospital, Beijing, China
| | - Aihua Cao
- Department of Pediatrics, Shandong University Qilu Hospital, Shandong, Jinan, China
| |
Collapse
|
49
|
Lesurf R, Breckpot J, Bouwmeester J, Hanafi N, Jain A, Liang Y, Papaz T, Lougheed J, Mondal T, Alsalehi M, Altamirano-Diaz L, Oechslin E, Audain E, Dombrowsky G, Postma AV, Woudstra OI, Bouma BJ, Hitz MP, Bezzina CR, Blue GM, Winlaw DS, Mital S. A validated heart-specific model for splice-disrupting variants in childhood heart disease. Genome Med 2024; 16:119. [PMID: 39402625 PMCID: PMC11476204 DOI: 10.1186/s13073-024-01383-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Congenital heart disease (CHD) is the most common congenital anomaly. Almost 90% of isolated cases have an unexplained genetic etiology after clinical testing. Non-canonical splice variants that disrupt mRNA splicing through the loss or creation of exon boundaries are not routinely captured and/or evaluated by standard clinical genetic tests. Recent computational algorithms such as SpliceAI have shown an ability to predict such variants, but are not specific to cardiac-expressed genes and transcriptional isoforms. METHODS We used genome sequencing (GS) (n = 1101 CHD probands) and myocardial RNA-Sequencing (RNA-Seq) (n = 154 CHD and n = 43 cardiomyopathy probands) to identify and validate splice disrupting variants, and to develop a heart-specific model for canonical and non-canonical splice variants that can be applied to patients with CHD and cardiomyopathy. Two thousand five hundred seventy GS samples from the Medical Genome Reference Bank were analyzed as healthy controls. RESULTS Of 8583 rare DNA splice-disrupting variants initially identified using SpliceAI, 100 were associated with altered splice junctions in the corresponding patient myocardium affecting 95 genes. Using strength of myocardial gene expression and genome-wide DNA variant features that were confirmed to affect splicing in myocardial RNA, we trained a machine learning model for predicting cardiac-specific splice-disrupting variants (AUC 0.86 on internal validation). In a validation set of 48 CHD probands, the cardiac-specific model outperformed a SpliceAI model alone (AUC 0.94 vs 0.67 respectively). Application of this model to an additional 947 CHD probands with only GS data identified 1% patients with canonical and 11% patients with non-canonical splice-disrupting variants in CHD genes. Forty-nine percent of predicted splice-disrupting variants were intronic and > 10 bp from existing splice junctions. The burden of high-confidence splice-disrupting variants in CHD genes was 1.28-fold higher in CHD cases compared with healthy controls. CONCLUSIONS A new cardiac-specific in silico model was developed using complementary GS and RNA-Seq data that improved genetic yield by identifying a significant burden of non-canonical splice variants associated with CHD that would not be detectable through panel or exome sequencing.
Collapse
Affiliation(s)
- Robert Lesurf
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jeroen Breckpot
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Jade Bouwmeester
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Nour Hanafi
- The Centre for Computational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anjali Jain
- The Centre for Computational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yijing Liang
- The Centre for Computational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Tanya Papaz
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Jane Lougheed
- Division of Cardiology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Tapas Mondal
- Division of Cardiology, Department of Pediatrics, McMaster Children's Hospital, Hamilton, ON, Canada
| | - Mahmoud Alsalehi
- Division of Cardiology, Department of Pediatrics, Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Luis Altamirano-Diaz
- Division of Cardiology, Department of Pediatrics, London Health Sciences Centre, London, ON, Canada
| | - Erwin Oechslin
- Division of Cardiology, Department of Medicine, Toronto Adult Congenital Heart Disease Program at Peter Munk Cardiac Centre, University Health Network, and University of Toronto, Toronto, ON, Canada
| | - Enrique Audain
- Institute of Medical Genetics, University Medicine Oldenburg, Carl von Ossietzky University, Oldenburg, Germany
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Kiel, Germany
| | - Gregor Dombrowsky
- Institute of Medical Genetics, University Medicine Oldenburg, Carl von Ossietzky University, Oldenburg, Germany
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Alex V Postma
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Odilia I Woudstra
- Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Berto J Bouma
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Marc-Phillip Hitz
- Institute of Medical Genetics, University Medicine Oldenburg, Carl von Ossietzky University, Oldenburg, Germany
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Kiel, Germany
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Gillian M Blue
- Heart Centre for Children, The Children's Hospital at Westmead, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - David S Winlaw
- Heart Center, Ann and Robert H. Lurie Children's Hospital of Chicago and Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Seema Mital
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.
- Ted Rogers Centre for Heart Research, Toronto, ON, Canada.
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
50
|
Dhawan A, Baitamouni S, Liu D, Eng C. Clinical Neurologic Features and Evaluation of PTEN Hamartoma Tumor Syndrome: A Systematic Review. Neurology 2024; 103:e209844. [PMID: 39250745 DOI: 10.1212/wnl.0000000000209844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND AND OBJECTIVES PTEN hamartoma tumor syndrome (PHTS) is a well-recognized hereditary tumor syndrome and is now also recognized as a common cause of monogenic autism spectrum disorder. There is a vast spectrum of phenotypic variability across individuals with PHTS, and in addition to neurodevelopmental challenges, patients with PHTS may experience a wide variety of neurologic challenges, many of which have only recently been described. Thus, this systematic review aimed to summarize the breadth of the current knowledge of neurologic conditions in individuals with PHTS. METHODS We conducted a systematic review using the MEDLINE and EMBASE databases until January 2023. We included studies that reported neurologic signs, symptoms, and diagnoses in patients with a diagnosis of PHTS. Two independent reviewers extracted data (neurologic diagnoses and patient details) from each study. Case reports, case series, prospective studies, and therapeutic trials were included. We assessed the quality of evidence using the appropriate tool from the JBI, depending on study design. RESULTS One thousand nine hundred ninety-six articles were screened, and 90 articles met the inclusion criteria. The majority of the included studies were case reports (49/90, 54%) or small case series (31/90, 34%). Epilepsy secondary to cerebral malformations, neurologic deficits from spinal or cranial arteriovenous malformations, and rare tumors such as dysplastic cerebellar gangliocytoma are among the more severe neurologic features reported across patients with PHTS. One interventional randomized control trial examining neurocognitive endpoints was identified and did not meet its efficacy endpoint. DISCUSSION Our systematic review defines a broad scope of neurologic comorbidities occurring in individuals with PHTS. Neurologic findings can be categorized by age at onset in individuals with PTHS. Our study highlights the need for additional clinical trial endpoints, informed by the neurologic challenges faced by individuals with PHTS.
Collapse
Affiliation(s)
- Andrew Dhawan
- From the Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, OH
| | - Sarah Baitamouni
- From the Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, OH
| | - Darren Liu
- From the Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, OH
| | - Charis Eng
- From the Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, OH
| |
Collapse
|