101
|
Pollak RM, Mortillo M, Murphy MM, Mulle JG. Behavioral Phenotypes and Comorbidity in 3q29 Deletion Syndrome: Results from the 3q29 Registry. J Autism Dev Disord 2025; 55:667-677. [PMID: 38216835 PMCID: PMC11814048 DOI: 10.1007/s10803-023-06218-w] [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] [Accepted: 12/06/2023] [Indexed: 01/14/2024]
Abstract
3q29 deletion syndrome (3q29del) is associated with a significantly increased risk for neurodevelopmental and neuropsychiatric disorders. However, the full spectrum of behavioral phenotypes associated with 3q29del is still evolving. Individuals with 3q29del (n = 96, 60.42% male) or their guardian completed the Achenbach Child or Adult Behavior Checklist (CBCL/ABCL) via the online 3q29 registry (3q29deletion.org). Typically developing controls (n = 57, 49.12% male) were ascertained as a comparison group. We analyzed mean performance on the CBCL/ABCL for individuals with 3q29del and controls across composite, DSM-keyed, and developmental scales; and the relationship between CBCL/ABCL performance and clinical and developmental phenotypes for individuals with 3q29del. Individuals with 3q29del showed significantly elevated behavioral and developmental impairment relative to controls across CBCL/ABCL domains. A substantial proportion of study participants with 3q29del scored in the Borderline or Clinical range for composite and DSM-keyed scales, indicating significant behavioral problems that may require clinical evaluation. We found that the preschool CBCL DSM-keyed autism spectrum problems scale is a potential screening tool for autism spectrum disorder (ASD) for individuals with 3q29del; CBCL/ABCL DSM-keyed scales were not accurate screeners for anxiety disorders or attention-deficit/hyperactivity disorder (ADHD) in our study sample. We identified a high degree of psychiatric comorbidity in individuals with 3q29del, with 60.42% (n = 58) of individuals with 3q29del scoring in the Borderline or Clinical range on two or more DSM-keyed CBCL/ABCL scales. Finally, we found that the degree of developmental delay in participants with 3q29del does not explain the increased behavioral problems observed on the CBCL/ABCL. The CBCL/ABCL can be used as screening tools in populations such as 3q29del, even in the presence of substantial psychiatric comorbidity. These results expand our understanding of the phenotypic spectrum of 3q29del and demonstrate an effective method for recruiting and phenotyping a large sample of individuals with a rare genetic disorder.
Collapse
Affiliation(s)
- Rebecca M Pollak
- Department of Psychiatry, Center for Advanced Biotechnology and Medicine, Robert Wood Johnson Medical School, Rutgers University, 679 Hoes Ln W, Piscataway, NJ, 08854, USA
| | - Michael Mortillo
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Melissa M Murphy
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Jennifer G Mulle
- Department of Psychiatry, Center for Advanced Biotechnology and Medicine, Robert Wood Johnson Medical School, Rutgers University, 679 Hoes Ln W, Piscataway, NJ, 08854, USA.
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
102
|
Vartiainen E, Liyanage D, Mazureac I, Battaglia RA, Tegtmeyer M, He JX, Räsänen N, Sealock J, McCarroll S, Nehme R, Pietiläinen O. Astrocytic-supplied cholesterol drives synaptic gene expression programs in developing neurons and downstream astrocytic transcriptional programs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635252. [PMID: 39975161 PMCID: PMC11838310 DOI: 10.1101/2025.01.28.635252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Astrocytes participate in neuronal synaptic programs that are enriched for genetic associations in schizophrenia and autism spectrum disorders (ASD). To better understand how these co-regulated cellular programs are induced during early neuronal development, we studied astrocytes and iPSC-derived neurons in co-cultures and mono-cultures at 16 time points spanning 0.5 hours to 8 days. We found that upregulation in astrocytes of genes involved in cholesterol biosynthesis preceded the activation of synaptic gene programs in neurons and upregulation of the astrocytic Nrxn1. Neuronal knockdown of key cholesterol receptors led to downregulation of neuronal synaptic genes and induced a robust transcriptional response in the astrocytes, including further upregulation of Nrxn1. This suggests that astrocyte-supplied cholesterol drives these neuronal changes and that bi-directional signalling is occuring. The genes upregulated in neurons were enriched for deleterious variants in schizophrenia and neurodevelopmental disorders, suggesting that their pathogenic effect may be, in part, mediated by reduced buffering capacity for changes in the astrocyte cholesterol supply to neurons. These findings highlight the critical role of astrocyte-neuron interactions in psychiatric and neurodevelopmental disorders, particularly in relation to lipid metabolism and synaptic plasticity.
Collapse
Affiliation(s)
- Emilia Vartiainen
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Dhara Liyanage
- The Stanley Center for Psychiatric Research at the Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Illinca Mazureac
- The Stanley Center for Psychiatric Research at the Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Rachel A Battaglia
- The Stanley Center for Psychiatric Research at the Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Matthew Tegtmeyer
- The Stanley Center for Psychiatric Research at the Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jax Xu He
- The Stanley Center for Psychiatric Research at the Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Noora Räsänen
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Julia Sealock
- The Stanley Center for Psychiatric Research at the Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Steven McCarroll
- The Stanley Center for Psychiatric Research at the Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Ralda Nehme
- The Stanley Center for Psychiatric Research at the Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Olli Pietiläinen
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- The Stanley Center for Psychiatric Research at the Broad Institute of Harvard and MIT, Cambridge, MA, USA
| |
Collapse
|
103
|
Porter RS, An S, Gavilan MC, Nagai M, Murata-Nakamura Y, Zhou B, Bonefas KM, Dionne O, Manuel JM, St-Germain J, Gascon S, Kim J, Browning L, Laurent B, Cho US, Iwase S. Coordinated neuron-specific splicing events restrict nucleosome engagement of the LSD1 histone demethylase complex. Cell Rep 2025; 44:115213. [PMID: 39817906 PMCID: PMC11864812 DOI: 10.1016/j.celrep.2024.115213] [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: 07/03/2023] [Revised: 10/15/2024] [Accepted: 12/24/2024] [Indexed: 01/18/2025] Open
Abstract
Chromatin regulatory proteins are expressed broadly and assumed to exert the same intrinsic function across cell types. Here, we report that 14 chromatin regulators undergo evolutionary-conserved neuron-specific splicing events involving microexons. Among them are two components of a histone demethylase complex: LSD1 H3K4 demethylase and the H3K4me0-reader PHF21A. We found that neuronal LSD1 splicing reduces the enzymes' affinity to the nucleosome. Meanwhile, neuronal PHF21A splicing significantly attenuates histone H3 binding and further ablates the DNA-binding function exerted by an AT-hook motif. Furthermore, in vitro reconstitution of the canonical and neuronal PHF21A-LSD1 complexes, combined with in vivo methylation mapping, identified the neuronal complex as a hypomorphic H3K4 demethylating machinery. The neuronal PHF21A, albeit with its weaker nucleosome binding, is necessary for normal gene expression and the H3K4 landscape in the developing brain. Thus, ubiquitously expressed chromatin regulatory complexes can exert neuron-specific functions via alternative splicing of their subunits.
Collapse
Affiliation(s)
- Robert S Porter
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sojin An
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria C Gavilan
- Genetics and Genomics Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Masayoshi Nagai
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yumie Murata-Nakamura
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Bo Zhou
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Katherine M Bonefas
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Olivier Dionne
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jeru Manoj Manuel
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Joannie St-Germain
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Suzanne Gascon
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Jacqueline Kim
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Liam Browning
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Benoit Laurent
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Uhn-Soo Cho
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Shigeki Iwase
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
104
|
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
|
105
|
Clémot-Dupont S, Lourenço Fernandes JA, Larrigan S, Sun X, Medisetti S, Stanley R, El Hankouri Z, Joshi SV, Picketts DJ, Shekhar K, Mattar P. The chromatin remodeler ADNP regulates neurodevelopmental disorder risk genes and neocortical neurogenesis. Proc Natl Acad Sci U S A 2025; 122:e2405981122. [PMID: 39808658 PMCID: PMC11760920 DOI: 10.1073/pnas.2405981122] [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: 03/23/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Although chromatin remodelers are among the most important risk genes associated with neurodevelopmental disorders (NDDs), the roles of these complexes during brain development are in many cases unclear. Here, we focused on the recently discovered ChAHP chromatin remodeling complex. The zinc finger and homeodomain transcription factor ADNP is a core subunit of this complex, and de novo ADNP mutations lead to intellectual disability and autism spectrum disorder. However, germline Adnp knockout mice were previously shown to exhibit early embryonic lethality, obscuring subsequent roles for the ChAHP complex in neurogenesis. To circumvent this early developmental arrest, we generated a conditional Adnp mutant allele. Using single-cell transcriptomics, cut&run-seq, and histological approaches, we show that during neocortical development, Adnp orchestrates the production of late-born, upper-layer neurons through a two-step process. First, Adnp is required to sustain progenitor proliferation specifically during the developmental window for upper-layer cortical neurogenesis. Accordingly, we found that Adnp recruits the ChAHP subunit Chd4 to genes associated with progenitor proliferation. Second, in postmitotic differentiated neurons, we define a network of risk genes linked to NDDs that are regulated by Adnp and Chd4. Taken together, these data demonstrate that ChAHP is critical for driving the expansion of upper-layer cortical neurons and for regulating neuronal gene expression programs, suggesting that these processes may potentially contribute to NDD etiology.
Collapse
Affiliation(s)
- Samuel Clémot-Dupont
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ONK1H 8L6
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ONK1H 8M5
| | - José Alex Lourenço Fernandes
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ONK1H 8L6
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ONK1H 8M5
| | - Sarah Larrigan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ONK1H 8L6
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ONK1H 8M5
| | - Xiaoqi Sun
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA94720
| | - Suma Medisetti
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ONK1H 8L6
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ONK1H 8M5
| | - Rory Stanley
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ONK1H 8L6
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ONK1H 8M5
| | - Ziyad El Hankouri
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ONK1H 8L6
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ONK1H 8M5
| | - Shrilaxmi V. Joshi
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ONK1H 8L6
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ONK1H 8M5
| | - David J. Picketts
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ONK1H 8L6
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ONK1H 8M5
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA94720
- Helen Wills Neuroscience Institute, Vision Science Graduate Group, Center for Computational Biology, Biophysics Graduate Group, California Institute of Quantitative Biosciences (QB3), University of California, Berkeley, CA94720
- Faculty Scientist, Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA94720
| | - Pierre Mattar
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ONK1H 8L6
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ONK1H 8M5
| |
Collapse
|
106
|
Collins RL, Talkowski ME. Diversity and consequences of structural variation in the human genome. Nat Rev Genet 2025:10.1038/s41576-024-00808-9. [PMID: 39838028 DOI: 10.1038/s41576-024-00808-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 01/23/2025]
Abstract
The biomedical community is increasingly invested in capturing all genetic variants across human genomes, interpreting their functional consequences and translating these findings to the clinic. A crucial component of this endeavour is the discovery and characterization of structural variants (SVs), which are ubiquitous in the human population, heterogeneous in their mutational processes, key substrates for evolution and adaptation, and profound drivers of human disease. The recent emergence of new technologies and the remarkable scale of sequence-based population studies have begun to crystalize our understanding of SVs as a mutational class and their widespread influence across phenotypes. In this Review, we summarize recent discoveries and new insights into SVs in the human genome in terms of their mutational patterns, population genetics, functional consequences, and impact on human traits and disease. We conclude by outlining three frontiers to be explored by the field over the next decade.
Collapse
Affiliation(s)
- Ryan L Collins
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael E Talkowski
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
107
|
Vaissiere T, Michaelson SD, Creson T, Goins J, Fürth D, Balazsfi D, Rojas C, Golovin R, Meletis K, Miller CA, O'Connor D, Fontolan L, Rumbaugh G. Syngap1 promotes cognitive function through regulation of cortical sensorimotor dynamics. Nat Commun 2025; 16:812. [PMID: 39827187 PMCID: PMC11743135 DOI: 10.1038/s41467-025-56125-0] [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: 10/30/2023] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Perception, a cognitive construct, emerges through sensorimotor integration (SMI). The genetic mechanisms that shape SMI required for perception are unknown. Here, we demonstrate in mice that expression of the autism/intellectual disability gene, Syngap1, in cortical excitatory neurons is required for the formation of somatomotor networks that promote SMI-mediated perception. Cortical Syngap1 expression was necessary and sufficient for setting tactile sensitivity, sustaining tactile object exploration, and promoting tactile learning. Mice with deficient Syngap1 expression exhibited impaired neural dynamics induced by exploratory touches within a cortical-thalamic network that promotes attention and perception. Disrupted neuronal dynamics were associated with circuit-specific long-range synaptic connectivity abnormalities. Our data support a model where autonomous Syngap1 expression in cortical excitatory neurons promotes cognitive abilities through the assembly of long-range circuits that integrate temporally-overlapping sensory and motor signals, a process that promotes perception and attention. These data provide systems-level insights into the robust association between Syngap1 expression and cognitive ability.
Collapse
Affiliation(s)
- Thomas Vaissiere
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Sheldon D Michaelson
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Thomas Creson
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Jessie Goins
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Daniel Fürth
- SciLifeLab, Department of Immunology, Genetics & Pathology, Uppsala University, Uppsala, Sweden
| | - Diana Balazsfi
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Camilo Rojas
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Randall Golovin
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | | | - Courtney A Miller
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL, USA
| | - Daniel O'Connor
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lorenzo Fontolan
- Aix-Marseille Université, INSERM, INMED, Turing Centre for Living Systems, Marseille, France
| | - Gavin Rumbaugh
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA.
| |
Collapse
|
108
|
Jung S, Caballero M, Olfson E, Newcorn JH, Fernandez TV, Mahjani B. Rare Variant Analyses in Ancestrally Diverse Cohorts Reveal Novel ADHD Risk Genes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.14.25320294. [PMID: 39867378 PMCID: PMC11759603 DOI: 10.1101/2025.01.14.25320294] [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: 01/28/2025]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly heritable neurodevelopmental disorder, but its genetic architecture remains incompletely characterized. Rare coding variants, which can profoundly impact gene function, represent an underexplored dimension of ADHD risk. In this study, we analyzed large-scale DNA sequencing datasets from ancestrally diverse cohorts and observed significant enrichment of rare protein-truncating and deleterious missense variants in highly evolutionarily constrained genes. This analysis identified 15 high-confidence ADHD risk genes, including the previously implicated KDM5B. Integrating these findings with genome-wide association study (GWAS) data revealed nine enriched pathways, with strong involvement in synapse organization, neuronal development, and chromatin regulation. Protein-protein interaction analyses identified chromatin regulators as central network hubs, and single-cell transcriptomic profiling confirmed their expression in neurons and glial cells, with distinct patterns in oligodendrocyte subtypes. These findings advance our understanding of the genetic architecture of ADHD, uncover core molecular mechanisms, and provide promising directions for future therapeutic development.
Collapse
Affiliation(s)
- Seulgi Jung
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madison Caballero
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily Olfson
- Child Study Center, Yale University, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Jeffrey H. Newcorn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas V. Fernandez
- Child Study Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Behrang Mahjani
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
109
|
Shih YT, Alipio JB, Klaft ZJ, Green N, Wong LP, Sadreyev R, Hyun JH, Dulla C, Sahay A. Pro-cognitive restoration of experience-dependent parvalbumin inhibitory neuron plasticity in neurodevelopmental disorders. RESEARCH SQUARE 2025:rs.3.rs-5624085. [PMID: 39877097 PMCID: PMC11774442 DOI: 10.21203/rs.3.rs-5624085/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
The hippocampus forms memories of our experiences by registering processed sensory information in coactive populations of excitatory principal cells or ensembles1-3. Fast-spiking parvalbumin-expressing inhibitory neurons (PV INs) in the dentate gyrus (DG)-CA3/CA2 circuit contribute to memory encoding by exerting precise temporal control of excitatory principal cell activity through mossy fiber-dependent feed-forward inhibition4-13. PV INs respond to input-specific information by coordinating changes in their intrinsic excitability, input-output synaptic-connectivity, synaptic-physiology and synaptic-plasticity9,13-17, referred to here as experience-dependent PV IN plasticity, to influence hippocampal functions. PV IN impairments in early life, when neural circuitry is highly sensitive to experience-dependent refinement, are thought to result in imbalanced excitation and inhibition, impaired cognition, network hyperexcitability and seizures: hallmarks of neurodevelopmental disorders (NDDs) such as Autism Spectrum Disorder and epilepsy18-20. Discovery of transcriptional regulators of experience-dependent PV IN plasticity in the adult hippocampus may permit reversal of these developmental impairments. Here, in a screen designed to capture the PV IN intrinsic program induced by increased mossy fiber inputs, a trigger for experience-dependent PV IN plasticity, we identify the homeobox gene Meis2 21 as a regulator of experience-dependent PV IN plasticity gene (XPG) in the adult DG-CA3/CA2 circuit. We found that a significant number of upregulated XPGs also exhibit haploinsufficiency in ASDs, epilepsies, and schizophrenia. We demonstrate that virally-mediated rescue of experience-dependent Meis2 upregulation in CA3/CA2 PV INs in a NDD risk mouse model in adulthood is sufficient to restore experience-dependent PV IN plasticity, spatial and social memory, ensemble specificity, suppression of network hyperexcitability and seizures. Together, these findings suggest that experience-dependent PV IN plasticity is a convergent mechanism for NDD risk genes that can be re-instated in adulthood to reverse developmental deficits in circuitry, network excitability and cognition.
Collapse
Affiliation(s)
- Yu-Tzu Shih
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jason Bondoc Alipio
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Zin-Juan Klaft
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Nathaniel Green
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Lai Ping Wong
- Department of Molecular Biology. Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology. Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jung Ho Hyun
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Chris Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
110
|
Medyanik AD, Anisimova PE, Kustova AO, Tarabykin VS, Kondakova EV. Developmental and Epileptic Encephalopathy: Pathogenesis of Intellectual Disability Beyond Channelopathies. Biomolecules 2025; 15:133. [PMID: 39858526 PMCID: PMC11763800 DOI: 10.3390/biom15010133] [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: 12/06/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Developmental and epileptic encephalopathies (DEEs) are a group of neuropediatric diseases associated with epileptic seizures, severe delay or regression of psychomotor development, and cognitive and behavioral deficits. What sets DEEs apart is their complex interplay of epilepsy and developmental delay, often driven by genetic factors. These two aspects influence one another but can develop independently, creating diagnostic and therapeutic challenges. Intellectual disability is severe and complicates potential treatment. Pathogenic variants are found in 30-50% of patients with DEE. Many genes mutated in DEEs encode ion channels, causing current conduction disruptions known as channelopathies. Although channelopathies indeed make up a significant proportion of DEE cases, many other mechanisms have been identified: impaired neurogenesis, metabolic disorders, disruption of dendrite and axon growth, maintenance and synapse formation abnormalities -synaptopathies. Here, we review recent publications on non-channelopathies in DEE with an emphasis on the mechanisms linking epileptiform activity with intellectual disability. We focus on three major mechanisms of intellectual disability in DEE and describe several recently identified genes involved in the pathogenesis of DEE.
Collapse
Affiliation(s)
- Alexandra D. Medyanik
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (A.D.M.); (P.E.A.); (A.O.K.); (E.V.K.)
| | - Polina E. Anisimova
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (A.D.M.); (P.E.A.); (A.O.K.); (E.V.K.)
| | - Angelina O. Kustova
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (A.D.M.); (P.E.A.); (A.O.K.); (E.V.K.)
| | - Victor S. Tarabykin
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (A.D.M.); (P.E.A.); (A.O.K.); (E.V.K.)
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Elena V. Kondakova
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (A.D.M.); (P.E.A.); (A.O.K.); (E.V.K.)
| |
Collapse
|
111
|
Kostyanovskaya E, Lasser MC, Wang B, Schmidt J, Bader E, Buteo C, Arbelaez J, Sindledecker AR, McCluskey KE, Castillo O, Wang S, Dea J, Helde KA, Graglia JM, Brimble E, Kastner DB, Ehrlich AT, State MW, Willsey AJ, Willsey HR. Convergence of autism proteins at the cilium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.05.626924. [PMID: 39677731 PMCID: PMC11643032 DOI: 10.1101/2024.12.05.626924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Hundreds of high-confidence autism genes have been identified, yet the relevant etiological mechanisms remain unclear. Gene ontology analyses have repeatedly identified enrichment of proteins with annotated functions in gene expression regulation and neuronal communication. However, proteins are often pleiotropic and these annotations are inherently incomplete. Our recent autism functional genetics work has suggested that these genes may share a common mechanism at the cilium, a membrane-bound organelle critical for neurogenesis, brain patterning, and neuronal activity-all processes strongly implicated in autism. Moreover, autism commonly co-occurs with conditions that are known to involve ciliary-related pathologies, including congenital heart disease, hydrocephalus, and blindness. However, the role of autism genes at the cilium has not been systematically investigated. Here we demonstrate that autism proteins spanning disparate functional annotations converge in expression, localization, and function at cilia, and that patients with pathogenic variants in these genes have cilia-related co-occurring conditions and biomarkers of disrupted ciliary function. This degree of convergence among genes spanning diverse functional annotations strongly suggests that cilia are relevant to autism, as well as to commonly co-occurring conditions, and that this organelle should be explored further for therapeutic potential.
Collapse
Affiliation(s)
- Elina Kostyanovskaya
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Micaela C. Lasser
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Belinda Wang
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - James Schmidt
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Ethel Bader
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Chad Buteo
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Juan Arbelaez
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Aria Rani Sindledecker
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Kate E. McCluskey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Octavio Castillo
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Sheng Wang
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Jeanselle Dea
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | | | | | | | - David B. Kastner
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Aliza T. Ehrlich
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Matthew W. State
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - A. Jeremy Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Helen Rankin Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub – San Francisco, San Francisco, CA
| |
Collapse
|
112
|
Wu XR, Yang L, Wu BS, Liu WS, Deng YT, Kang JJ, Dong Q, Sahakian BJ, Feng JF, Cheng W, Yu JT. Exome sequencing identifies genes for socioeconomic status in 350,770 individuals. Proc Natl Acad Sci U S A 2025; 122:e2414018122. [PMID: 39772748 PMCID: PMC11745334 DOI: 10.1073/pnas.2414018122] [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: 07/12/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Socioeconomic status (SES) is a critical factor in determining health outcomes and is influenced by genetic and environmental factors. However, our understanding of the genetic structure of SES remains incomplete. Here, we conducted a large-scale exome study of SES markers (household income, occupational status, educational attainment, and social deprivation) in 350,770 individuals. For rare coding variants, we identified 56 significant associations by gene-based collapsing tests, unveiling 7 additional SES-associated genes (NRN1, CCDC36, RHOB, EP400, NCAM1, TPTEP2-CSNK1E, and LINC02881). Exome-wide single common variant analysis revealed nine lead single-nucleotide polymorphisms (SNPs) associated with household income and 34 lead SNPs associated with EduYears, replicating previous GWAS findings. The gene-environment correlations had a substantial impact on the genetic associations with SES, as indicated by the significantly increased P values in several associations after controlling for geographic regions. Furthermore, we observed the pleiotropic effects of SES-associated genetic factors on a wide range of health outcomes, such as cognitive function, psychosocial status, and diabetes. This study highlights the contribution of coding variants to SES and their associations with health phenotypes.
Collapse
Affiliation(s)
- Xin-Rui Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai200433, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Barbara J. Sahakian
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai200433, China
- Department of Psychiatry and Behavioural and Clinical Neuroscience Institute, University of Cambridge, CambridgeCB2 0SZ, United Kingdom
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai200433, China
- Department of Computer Science, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai200433, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| |
Collapse
|
113
|
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
|
114
|
Mamat M, Chen Y, Shen W, Li L. Molecular architecture of the altered cortical complexity in autism. Mol Autism 2025; 16:1. [PMID: 39763008 PMCID: PMC11705879 DOI: 10.1186/s13229-024-00632-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Autism spectrum disorder (ASD) is characterized by difficulties in social interaction, communication challenges, and repetitive behaviors. Despite extensive research, the molecular mechanisms underlying these neurodevelopmental abnormalities remain elusive. We integrated microscale brain gene expression data with macroscale MRI data from 1829 participants, including individuals with ASD and typically developing controls, from the autism brain imaging data exchange I and II. Using fractal dimension as an index for quantifying cortical complexity, we identified significant regional alterations in ASD, within the left temporoparietal, left peripheral visual, right central visual, left somatomotor (including the insula), and left ventral attention networks. Partial least squares regression analysis revealed gene sets associated with these cortical complexity changes, enriched for biological functions related to synaptic transmission, synaptic plasticity, mitochondrial dysfunction, and chromatin organization. Cell-specific analyses, protein-protein interaction network analysis and gene temporal expression profiling further elucidated the dynamic molecular landscape associated with these alterations. These findings indicate that ASD-related alterations in cortical complexity are closely linked to specific genetic pathways. The combined analysis of neuroimaging and transcriptomic data enhances our understanding of how genetic factors contribute to brain structural changes in ASD.
Collapse
Affiliation(s)
- Makliya Mamat
- School of Basic Medical Sciences, Health Science Center, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Yiyong Chen
- School of Basic Medical Sciences, Health Science Center, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo, 315211, Zhejiang, People's Republic of China.
| | - Wenwen Shen
- Affiliated Kangning Hospital of Ningbo University, Ningbo, 315201, Zhejiang, People's Republic of China.
| | - Lin Li
- Human Anatomy Department, Nanjing Medical University, No.101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, People's Republic of China.
| |
Collapse
|
115
|
Lee KS, Lee T, Kim M, Ignatova E, Ban HJ, Sung MK, Kim Y, Kim YJ, Han JH, Choi JK. Shared rare genetic variants in multiplex autism families suggest a social memory gene under selection. Sci Rep 2025; 15:696. [PMID: 39753649 PMCID: PMC11698849 DOI: 10.1038/s41598-024-83839-w] [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: 10/02/2023] [Accepted: 12/17/2024] [Indexed: 01/06/2025] Open
Abstract
Autism spectrum disorder (ASD) affects up to 1 in 59 children, and is one of the most common neurodevelopmental disorders. Recent genomic studies have highlighted the role of rare variants in ASD. This study aimed to identify genes affected by rare variants shared by siblings with ASD and validate the function of a candidate gene FRRS1L. By integrating the whole genome sequencing data of 866 multiplex families from the Hartwell Foundation's Autism Research and Technology Initiative and Autism Speaks MSSNG project, we identified rare variants shared by two or more siblings with ASD. Using shared rare variants (SRVs), we selected candidate genes for ASD. Gene prioritization by evolutionary features and expression alterations on autism identified FRRS1L in two families, including one with impaired social behaviors. One variant in this family was 6 bp away from human-specific trinucleotide fixation. Additionally, CRISPR/Cas9 experiments demonstrated downregulation by a family variant and upregulation by a fixed site. Population genetics further demonstrated that upregulation of this gene has been favored during human evolution. Various mouse behavioral tests showed that Frrs1l knockout specifically impairs social novelty recognition without altering other behavioral phenotypes. Furthermore, we constructed humanized mice by introducing human sequences into a mouse genome. These knockin mice showed improved abilities to retain social memory over time. The results of our population genetic and evolutionary analyses, behavior experiments, and genome editing propose a molecular mechanism that may confer a selective advantage through social memory enhancement and may cause autism-related social impairment when disrupted in humans. These findings highlight the role of FRRS1L, the AMPA receptor subunit, in social behavior and evolution.
Collapse
Affiliation(s)
- Kang Seon Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Taeyeop Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
- Department of Psychiatry, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
- Translational Biomedical Research Group, Asan Institute for Life Science, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Mujun Kim
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Elizaveta Ignatova
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Hyo-Jeong Ban
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Min Kyung Sung
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Younghoon Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Youn-Jae Kim
- Specific Organs Cancer Branch, National Cancer Center, Gyeonggi, 10408, Republic of Korea
| | - Jin-Hee Han
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea.
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
116
|
Luo S, Wang PY, Zhou P, Zhang WJ, Gu YJ, Liang XY, Zhang JW, Luo JX, Zhang HW, Lan S, Zhang TT, Yang JH, Sun SZ, Guo XY, Wang JL, Deng LF, Xu ZH, Jin L, He YY, Ye ZL, Gu WY, Li BM, Shi YW, Liu XR, Yan HJ, Yi YH, Jiang YW, Mao X, Li WL, Meng H, Liao WP. Variants in EP400, encoding a chromatin remodeler, cause epilepsy with neurodevelopmental disorders. Am J Hum Genet 2025; 112:87-105. [PMID: 39708813 PMCID: PMC11739926 DOI: 10.1016/j.ajhg.2024.11.010] [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: 06/18/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/23/2024] Open
Abstract
EP400 encodes a core catalytic ATPase subunit of ATP-dependent chromatin remodeling complexes. The gene-disease association of EP400 is undetermined. In this study, we performed trio-based whole-exome sequencing in a cohort of 402 families with epilepsy and neurodevelopmental disorders (NDDs) and identified compound heterozygous EP400 variants in six unrelated individuals. Six additional EP400 individuals were recruited via the match platform of China, including two de novo heterozygous and four compound heterozygous variants. The individual with a heterozygous de novo frameshift variant presented with NDDs, while the others exhibited epilepsy and NDDs, explained by the damaged genetic dependence quantity. EP400 presented significantly higher excesses of variants in the individuals. Clustering analysis revealed that the majority paralogs of EP400 were associated with NDDs/epilepsy and co-expressed highly with EP400. Analysis of the spatiotemporal expression indicated that EP400 is highly expressed in the developing brain and cells during differentiation, indicating its vital role in neurodevelopment; EP400 is predominantly expressed in inhibitory neurons in the early stage but in excitatory neurons in the mature stage. The development-dependent expression pattern of neuron specificity explained the favorable outcome of epilepsy. Knockdown of EP400 ortholog in Drosophila caused significantly increased susceptibility to seizures and abnormal neuronal firing. The ep400 crispant zebrafish exhibited brain developmental abnormalities, poorer adaptability, lower response to stimulation, epileptic discharges, abnormal cellular apoptosis, and increased susceptibility to seizures. Transcriptome analysis showed that ep400 deficiency caused expressional dysregulation of 84 epilepsy/NDD-associated genes, including 11 highly dose-sensitive genes. This study identified EP400 as a causative gene of epilepsy/NDDs.
Collapse
Affiliation(s)
- Sheng Luo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Peng-Yu Wang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Peng Zhou
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Wen-Jun Zhang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Yu-Jie Gu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Xiao-Yu Liang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Jing-Wen Zhang
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Jun-Xia Luo
- Epilepsy Center, Qilu Children's Hospital of Shandong University, Jinan 250000, Shandong, China
| | - Hong-Wei Zhang
- Epilepsy Center, Qilu Children's Hospital of Shandong University, Jinan 250000, Shandong, China
| | - Song Lan
- Department of Neurology, Maoming People's Hospital, Maoming 525000, Guangdong, China
| | - Ting-Ting Zhang
- Department of Psychology, Guangdong Sanjiu Brain Hospital, Guangzhou 510440, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jie-Hua Yang
- Department of Neurology, Second Affiliated Hospital of Shantou University, Shantou 515000, Guangdong, China
| | - Su-Zhen Sun
- Department of Neurology, Hebei Children's Hospital, Shijiazhuang 050000, Hebei, China
| | - Xiang-Yang Guo
- Department of Pediatrics, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Ju-Li Wang
- Epilepsy Center, Jiamusi Central Hospital, Jiamusi 154002, Heilongjiang, China
| | - Lin-Fan Deng
- Department of Pediatrics, Mianyang Central Hospital, Mianyang 621000, Sichuan, China
| | - Ze-Hai Xu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Liang Jin
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Yun-Yan He
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Zi-Long Ye
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Wei-Yue Gu
- Beijing Chigene Translational Medicine Research Center Co., Ltd., Beijing 100000, China
| | - Bing-Mei Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Yi-Wu Shi
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Xiao-Rong Liu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Hong-Jun Yan
- Epilepsy Center, Guangdong Sanjiu Brain Hospital, Guangzhou 510440, Guangdong, China
| | - Yong-Hong Yi
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China
| | - Yu-Wu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xiao Mao
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, China
| | - Wen-Ling Li
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Heng Meng
- Department of Neurology, the First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Avenue, Guangzhou, China.
| | - Wei-Ping Liao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, Guangdong, China.
| |
Collapse
|
117
|
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
|
118
|
Fujioka Y, Shiura H, Ishii M, Ono R, Endo T, Kiyonari H, Hirate Y, Ito H, Kanai-Azuma M, Kohda T, Kaneko-Ishino T, Ishino F. Targeting of retrovirus-derived Rtl8a/ 8b causes late-onset obesity, reduced social response and increased apathy-like behaviour. Open Biol 2025; 15:240279. [PMID: 39875098 PMCID: PMC11774587 DOI: 10.1098/rsob.240279] [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: 09/26/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 01/30/2025] Open
Abstract
Retrotransposon Gag-like (RTL) 8A, 8B and 8C are eutherian-specific genes derived from a certain retrovirus. They cluster as a triplet of genes on the X chromosome, but their function remains unknown. Here, we demonstrate that Rtl8a and Rtl8b play important roles in the brain: their double knockout (DKO) mice not only exhibit reduced social responses and increased apathy-like behaviour, but also become obese from young adulthood, similar to patients with late Prader-Willi syndrome (PWS), a neurodevelopmental genomic imprinting disorder. Mouse RTL8A/8B proteins are expressed in the prefrontal cortex and hypothalamus and localize to both the nucleus and cytoplasm of neurons, presumably due to the N-terminal nuclear localization signal-like sequence at the N-terminus. An RNAseq study in the cerebral cortex revealed reduced expression of several GABA type A receptor subunit genes in DKO, in particular Gabrb2, which encodes its β2 subunit. We confirmed the reduction of GABRB2 protein in the DKO cerebral cortex by western blotting. As GABRB2 has been implicated in the aetiology of several neurodevelopmental and neuropsychiatric disorders, it is likely that the reduction of GABRB2 is one of the major causes of the neuropsychiatric defects in the DKO mice.
Collapse
Affiliation(s)
- Yoshifumi Fujioka
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
| | - Hirosuke Shiura
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Faculty of Life and Environmental Sciences, University of Yamanashi, Kohfu,Yamanashi 400-8510, Japan
| | - Masayuki Ishii
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
| | - Ryuichi Ono
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences (NIHS), Kawasaki, Kanagawa210-9501, Japan
| | - Tsutomu Endo
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo650-0047, Japan
| | | | - Hikaru Ito
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
- Research Facility Center for Science and Technology, Kagawa University, Takamatsu,Kagawa 761-0793, Japan
| | | | - Takashi Kohda
- Faculty of Life and Environmental Sciences, University of Yamanashi, Kohfu,Yamanashi 400-8510, Japan
| | - Tomoko Kaneko-Ishino
- Faculty of Nursing, Tokai University School of Medicine, Isehara, Kanagawa259-1193, Japan
| | - Fumitoshi Ishino
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
| |
Collapse
|
119
|
Xie S, Zuo K, De Rubeis S, Ruggerone P, Carloni P. Molecular basis of the CYFIP2 and NCKAP1 autism-linked variants in the WAVE regulatory complex. Protein Sci 2025; 34:e5238. [PMID: 39660913 PMCID: PMC11632847 DOI: 10.1002/pro.5238] [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: 09/27/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024]
Abstract
The WAVE regulatory pentameric complex regulates actin remodeling. Two components of it (CYFIP2 and NCKAP1) are encoded by genes whose genetic mutations increase the risk for autism spectrum disorder (ASD) and related neurodevelopmental disorders. Here, we use a newly developed computational protocol and hotspot analysis to uncover the functional impact of these mutations at the interface of the correct isoforms of the two proteins into the complex. The mutations turn out to be located on the surfaces involving the largest number of hotspots of the complex. Most of them decrease the affinity of the proteins for the rest of the complex, but some have the opposite effect. The results are fully consistent with the available experimental data. The observed changes in the WAVE regulatory complex stability might impact on complex activation and ultimately play a role in the aberrant pathway of the complex, leading to the cell derangement associated with the disease.
Collapse
Affiliation(s)
- Song Xie
- Computational BiomedicineInstitute of Advanced Simulation IAS‐5 and Institute of Neuroscience and Medicine INM‐9, Forschungszentrum Jülich GmbHJülichGermany
- Department of PhysicsRWTH Aachen UniversityAachenGermany
| | - Ke Zuo
- Computational BiomedicineInstitute of Advanced Simulation IAS‐5 and Institute of Neuroscience and Medicine INM‐9, Forschungszentrum Jülich GmbHJülichGermany
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative MedicineCollege of Pharmacy (International Academy of Targeted Therapeutics and Innovation), Chongqing University of Arts and SciencesChongqingChina
- Department of PhysicsUniversity of CagliariMonserratoCagliariItaly
| | - Silvia De Rubeis
- Seaver Autism Center for Research and TreatmentIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- The Mindich Child Health and Development InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Paolo Ruggerone
- Department of PhysicsUniversity of CagliariMonserratoCagliariItaly
| | - Paolo Carloni
- Computational BiomedicineInstitute of Advanced Simulation IAS‐5 and Institute of Neuroscience and Medicine INM‐9, Forschungszentrum Jülich GmbHJülichGermany
- Department of PhysicsRWTH Aachen UniversityAachenGermany
- JARA Institute: Molecular Neuroscience and ImagingInstitute of Neuroscience and Medicine INM‐11, Forschungszentrum Jülich GmbHJülichGermany
| |
Collapse
|
120
|
Akter H, Rahaman MA, Eshaque TB, Mohamed N, Islam A, Morshed M, Shahin Z, Muhaimin A, Foyzullah AM, Mim RA, Omar FB, Hasan MN, Satsangi D, Ahmed N, Al Saba A, Jahan N, Hossen MA, Mondol MA, Sakib AS, Kabir R, Jahan Chowdhury MS, Shams N, Afroz S, Kanta SI, Bhuiyan SJ, Biswas R, Hanif S, Tambi R, Nassir N, Rahman MM, Duan J, D Børglum A, Amin R, Basiruzzaman M, Kamruzzaman M, Sarker S, Woodbury-Smith M, Uddin KMF, Nabi AHMN, Uddin M. Genomic insights from a deeply phenotyped highly consanguineous neurodevelopmental disorders cohort. Genet Med 2025; 27:101282. [PMID: 39342494 DOI: 10.1016/j.gim.2024.101282] [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: 03/25/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
PURPOSE The genetic underpinning of neurodevelopmental disorders (NDDs) in diverse ethnic populations, especially those with high rates of consanguinity, remains largely unexplored. Here, we aim to elucidate genomic insight from 576 well-phenotyped and highly consanguineous (16%) NDD cohort. METHODS We used chromosomal microarray (CMA; N:247), exome sequencing (ES; N:127), combined CMA and ES (N:202), and long-read genome sequencing to identify genetic etiology. Deep clinical multivariate data were coupled with genomic variants for stratification analysis. RESULTS Genetic diagnosis rates were 17% with CMA, 29.92% with ES, and 37.13% with combined CMA and ES. Notably, children of consanguineous parents showed a significantly higher diagnostic yield (P < .01) compared to those from nonconsanguineous parents. Among the ES-identified pathogenic variants, 36.19% (38/105) were novel, implicating 35 unique genes. Long-read sequencing of seizure participants unresolved by combined test identified expanded FMR1 trinucleotide repeats. Additionally, we identified 2 recurrent X-linked variants in the G6PD in 3.65% (12/329) of NDD participants. These variants were absent in large-population control cohorts and cohort comprising neurodevelopmental and neuropsychiatric populations of European descendants, indicating a possible associated risk factor potentially resulting from ancient genetic drift. CONCLUSION This study unveils unique clinical and genomic insights from a consanguinity rich Bangladeshi NDD cohort.
Collapse
Affiliation(s)
- Hosneara Akter
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh; Laboratory of Population Genetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Md Atikur Rahaman
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | | | - Nesrin Mohamed
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, UAE
| | - Amirul Islam
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh; GenomeArc Inc., Mississauga, ON, Canada
| | - Mehzabin Morshed
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Zaha Shahin
- Imperial College London, London, United Kingdom
| | - Al Muhaimin
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Arif Md Foyzullah
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Rabeya Akter Mim
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Farjana Binta Omar
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Md Nahid Hasan
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh; GenomeArc Inc., Mississauga, ON, Canada
| | - Dharana Satsangi
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Nahid Ahmed
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Abdullah Al Saba
- Laboratory of Population Genetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Nargis Jahan
- Centre for Precision Therapeutics (NPT), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Md Arif Hossen
- Centre for Precision Therapeutics (NPT), NeuroGen Healthcare, Dhaka, Bangladesh
| | | | | | - Rezwana Kabir
- Centre for Precision Therapeutics (NPT), NeuroGen Healthcare, Dhaka, Bangladesh
| | | | - Nusrat Shams
- Centre for Precision Therapeutics (NPT), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Shireen Afroz
- Bangladesh Shishu Hospital and Institute, Dhaka, Bangladesh
| | | | | | - Rabi Biswas
- Bangladesh Shishu Hospital and Institute, Dhaka, Bangladesh
| | - Shehzad Hanif
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, UAE
| | - Richa Tambi
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, UAE
| | - Nasna Nassir
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, UAE; College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Muhammad Mizanur Rahman
- Department of Paediatric Neurology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Jinjie Duan
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Department of Biomedicine and the iSEQ Centre, Aarhus University, Aarhus, Denmark; Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Anders D Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Department of Biomedicine and the iSEQ Centre, Aarhus University, Aarhus, Denmark; Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Robed Amin
- Ministry of Health and Family Welfare, Government of People Republic of Bangladesh, Dhaka, Bangaldesh
| | | | - Md Kamruzzaman
- Bangladesh Shishu Hospital and Institute, Dhaka, Bangladesh
| | - Shaoli Sarker
- Centre for Precision Therapeutics (NPT), NeuroGen Healthcare, Dhaka, Bangladesh; Bangladesh Shishu Hospital and Institute, Dhaka, Bangladesh
| | - Marc Woodbury-Smith
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - K M Furkan Uddin
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh; Department of Biochemistry, Holy Family Red Crescent Medical College, Dhaka, Bangladesh
| | - A H M Nurun Nabi
- Laboratory of Population Genetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Mohammed Uddin
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, UAE; GenomeArc Inc., Mississauga, ON, Canada; College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE.
| |
Collapse
|
121
|
O'Connell M, Harstad E, Aites J, Hayes K, Arnett AB, Scotellaro J, Patel S, Brewster SJ, Barbaresi W, Doan RN. Diverse clinical presentation of SPTBN1 variants: Complex versus primary attention-deficit/hyperactivity disorder. Am J Med Genet A 2025; 197:e63851. [PMID: 39162370 DOI: 10.1002/ajmg.a.63851] [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: 10/04/2023] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) belongs to a phenotypically broad class of mental health disorders impacting social and cognitive functioning. Despite heritability estimates of 77%-88% and a global prevalence of up to 1 in 20 children, most of the underlying genetic etiology of the disorder remains undiscovered, making it challenging to obtain a clinical molecular genetic diagnosis and to develop new treatments (Biological Psychiatry, 2005, 57, 1313; Psychological Bulletin, 2009, 135, 608; Psychological Medicine, 2014, 44, 2223). Here we report the identification of a novel ultra-rare heterozygous loss-of-function (p.Q1625*) variant in a child with complex ADHD (i.e., comorbid mild intellectual disability [ID]) and a missense (p.G1748R) variant (allele frequency of 4.7 × 10-5) in a child with primary ADHD (i.e., absence of comorbid autism spectrum disorder [ASD], ID, or syndromic features) both in the SPTBN1 gene. Missense variants in SPTBN1 have been reported in individuals with developmental disorders, language and communication disorders, and motor delays in recent publications (Nature Genetics, 2021, 53, 1006; American Journal of Medical Genetics Part A, 2021, 185, 2037) and ClinVar, though most variants in ClinVar have uncertain disease associations. The functional impact of these 135 variants, including from the current study, were further assessed using prediction scores from the recently developed AlphaMissense tool and benchmarked against published functional studies on a subset of the variants. While heterozygous SPTBN1 variants have recently been associated with neurodevelopmental disorders characterized by global developmental delay, intellectual disability, and behavioral abnormalities, the two patients in the current study expand the phenotypic spectrum to include ADHD in the absence of more severe neurodevelopmental disorders, such as ASD and moderate to severe ID. Furthermore, the culmination of these data with existing reported cases suggests that variation including loss of function and missense events underlie a broader clinical spectrum than previously understood.
Collapse
Affiliation(s)
- Mia O'Connell
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics & Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Elizabeth Harstad
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Developmental Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jennifer Aites
- Division of Developmental Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Katheryn Hayes
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics & Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Anne B Arnett
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Developmental Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Julia Scotellaro
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics & Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Soleha Patel
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics & Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Stephanie J Brewster
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics & Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
- Division of Developmental Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - William Barbaresi
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Developmental Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Ryan N Doan
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics & Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
122
|
Graglia JM, Harding AJ, Helde KA. Roadmap to advance therapeutics for SYNGAP1-related disorder: a patient organization perspective from SynGAP Research Fund. THERAPEUTIC ADVANCES IN RARE DISEASE 2025; 6:26330040241308285. [PMID: 39807402 PMCID: PMC11726535 DOI: 10.1177/26330040241308285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
SYNGAP1-related disorder (SRD) is a developmental and epileptic encephalopathy caused by a disruption of the SYNGAP1 gene. At the beginning of 2024, it is one of many rare monogenic brain disorders without disease-modifying treatments, but that is changing. This article chronicles the last 5 years, beginning when treatments for SRD were not publicly in development, to the start of 2024 when many SRD-specific treatments are advancing. We discuss the progress across many realms that have brought SRD to the forefront of drug development and highlight how Patient Advocacy Groups (PAGs) have had direct roles in accelerating the route to meaningful treatments for our children. We start with a summary of why SRD is an attractive pharmaceutical target. Second, we introduce the disease, the clinical features, and the number of patients. Next, we describe our PAG, our international partners and cite examples of the broad range of activities we believe are accelerating our pace toward treatments. We summarize the current SYNGAP1 pipeline and the status of each public project. Finally, we discuss two open questions that urgently need to be addressed in advance of clinical trials for SRD.
Collapse
|
123
|
Chalkiadaki K, Statoulla E, Zafeiri M, Voudouri G, Amvrosiadis T, Typou A, Theodoridou N, Moschovas D, Avgeropoulos A, Samiotaki M, Mason JO, Gkogkas CG. GABA/Glutamate Neuron Differentiation Imbalance and Increased AKT/mTOR Signaling in CNTNAP2 -/- Cerebral Organoids. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100413. [PMID: 39758604 PMCID: PMC11699409 DOI: 10.1016/j.bpsgos.2024.100413] [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: 07/30/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 01/07/2025] Open
Abstract
Background The polygenic nature of autism spectrum disorder (ASD) requires the identification of converging genetic pathways during early development to elucidate its complexity and varied manifestations. Methods We developed a human cerebral organoid model from induced pluripotent stem cells with targeted genome editing to abolish protein expression of the CNTNAP2 ASD risk gene. Results CNTNAP2-/- cerebral organoids displayed accelerated cell cycle, ventricular zone disorganization, and increased cortical folding. Proteomic analysis revealed disruptions in glutamatergic/GABAergic (gamma-aminobutyric acidergic) synaptic pathways and neurodevelopment, and transcriptomic analysis revealed differentially expressed genes belonging to inhibitory neuron-related gene networks. Interestingly, there was a weak correlation between the 2 datasets, suggesting nuanced translational control mechanisms. Along these lines, we found upregulated AKT/mTOR (mechanistic target of rapamycin) signaling in CNTNAP2-/- organoids. Spatial transcriptomic analysis of CNTNAP2-/- ventricular-like zones demonstrated pervasive changes in gene expression, implicating upregulation of cell cycle regulation, synaptic, and glutamatergic/GABAergic pathways. We noted significant overlap of all day-30 organoid omics datasets differentially expressed genes from idiopathic ASD (macrocephaly) induced pluripotent stem cell-derived telencephalic organoids, where FOXG1 was upregulated. Moreover, we detected increased GAD1-expressing and decreased TBR1-expressing cells, suggesting altered GABAergic/glutamatergic neuron development. Conclusions These findings potentially highlight a shared mechanism in the early cortical development of various forms of ASD, further elucidate the role of CNTNAP2 in ASD pathophysiology and cortical development, and pave the way for targeted therapies that use cerebral organoids as preclinical models.
Collapse
Affiliation(s)
- Kleanthi Chalkiadaki
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | - Elpida Statoulla
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | - Maria Zafeiri
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | - Georgia Voudouri
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | | | - Alexandra Typou
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | - Niki Theodoridou
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | - Dimitrios Moschovas
- Department of Materials Science Engineering, University of Ioannina, Ioannina, Greece
| | | | - Martina Samiotaki
- Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece
| | - John O. Mason
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, United Kingdom
| | - Christos G. Gkogkas
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| |
Collapse
|
124
|
Han X, He Y, Wang Y, Hu W, Chu C, Huang L, Hong Y, Han L, Zhang X, Gao Y, Lin Y, Ma H, Shen H, Ke X, Liu Y, Hu Z. Deficiency of FABP7 Triggers Premature Neural Differentiation in Idiopathic Normocephalic Autism Organoids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406849. [PMID: 39556706 PMCID: PMC11727249 DOI: 10.1002/advs.202406849] [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: 06/20/2024] [Revised: 10/30/2024] [Indexed: 11/20/2024]
Abstract
Autism spectrum disorder (ASD), which is caused by heterogeneous genetic and environmental factors, is characterized by diverse clinical phenotypes linked to distinct pathological mechanisms. ASD individuals with a shared clinical phenotype might contribute to revealing the molecular mechanism underlying ASD progression. Here, it is generated induced pluripotent stem cell (iPSC)-derived cerebral organoids from normocephalic individuals with ASD in a prospective birth cohort with a shared clinical diagnosis. Multiple cell lines and time series scRNA-seq combined with a histomorphological analysis revealed premature neural differentiation of neural stem cells (NSCs) and decreased expression of Fatty acid binding protein 7 (FABP7) in ASD organoids. It is subsequently revealed alterations in the phosphorylation levels of Mitogen-Activated Protein Kinase Kinase 1/2 (MEK1/2), which are downstream of FABP7, and the regulation of the FABP7/MEK pathway reversed improper neural differentiation in the ASD organoids. Moreover, both Fabp7-knockdown and MEK2-overexpressing mice exhibited repetitive stereotyped behaviors and social defects relevant to autism. This study reveals the role of the FABP7/MEK pathway in abnormal NSC differentiation in normocephalic individuals with ASD, which might provide a promising therapeutic target for ASD treatment.
Collapse
Affiliation(s)
- Xiao Han
- Interdisciplinary Inno Center for Organoids, State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjing211166China
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Innovation Center of SuzhouNanjing Medical UniversitySuzhou215000China
| | - Yuanlin He
- Interdisciplinary Inno Center for Organoids, State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Innovation Center of SuzhouNanjing Medical UniversitySuzhou215000China
- Department of Epidemiology and Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Yuanhao Wang
- Interdisciplinary Inno Center for Organoids, State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjing211166China
| | - Wenzhu Hu
- Department of Epidemiology and Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Chu Chu
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjing211166China
| | - Lei Huang
- Interdisciplinary Inno Center for Organoids, State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
- Department of Epidemiology and Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Yuan Hong
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjing211166China
| | - Lu Han
- Autism Research Center, State Key Laboratory of Reproductive MedicineThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjing210029China
| | - Xu Zhang
- Interdisciplinary Inno Center for Organoids, State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
| | - Yao Gao
- Department of Epidemiology and Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Yuan Lin
- Interdisciplinary Inno Center for Organoids, State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
- Department of Maternal, Child and Adolescent Health, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Hongxia Ma
- Interdisciplinary Inno Center for Organoids, State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
- Department of Epidemiology and Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Xiaoyan Ke
- Autism Research Center, State Key Laboratory of Reproductive MedicineThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjing210029China
| | - Yan Liu
- Interdisciplinary Inno Center for Organoids, State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjing211166China
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Innovation Center of SuzhouNanjing Medical UniversitySuzhou215000China
| | - Zhibin Hu
- Interdisciplinary Inno Center for Organoids, State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Innovation Center of SuzhouNanjing Medical UniversitySuzhou215000China
- Department of Epidemiology and Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjing211166China
| |
Collapse
|
125
|
Heine VM, Dooves S. Neuroglia in autism spectrum disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:303-311. [PMID: 40148051 DOI: 10.1016/b978-0-443-19102-2.00006-5] [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: 03/29/2025]
Abstract
Autism spectrum disorder (ASD) is characterized by difficulties in social interaction, communication, and repetitive behavior, typically diagnosed during early childhood and attributed to altered neuronal network connectivity. Several genetic and environmental risk factors contribute to ASD, including pre- or early life immune activation, which can trigger microglial and astroglial reactivity, impacting early neurodevelopment. In ASD, astrocytes show altered glutamate metabolism, directly influencing neuronal network activity, while microglia display impaired synaptic pruning, an essential developmental process for the refinement of neuronal connections. Additionally, reduced myelination in specific cortical and subcortical regions may affect brain connectivity in ASD, with white matter integrity correlating with the severity of the disorder, suggesting an important role for oligodendrocytes and myelin in ASD. This chapter provides an overview of current literature on the role of neuroglia cells in ASD, with a focus on immune activation, glutamate signaling, synaptic pruning, and myelination.
Collapse
Affiliation(s)
- Vivi M Heine
- Department of Child and Adolescence Psychiatry, Emma Center for Personalized Medicine, Amsterdam Neuroscience, Emma Children's Hospital, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Stephanie Dooves
- Department of Child and Adolescence Psychiatry, Emma Center for Personalized Medicine, Amsterdam Neuroscience, Emma Children's Hospital, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
126
|
Kong C, Bing Z, Yang L, Huang Z, Wang W, Grebogi C. Transcriptomic Evidence Reveals the Dysfunctional Mechanism of Synaptic Plasticity Control in ASD. Genes (Basel) 2024; 16:11. [PMID: 39858558 PMCID: PMC11764921 DOI: 10.3390/genes16010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND/OBJECTIVES A prominent endophenotype in Autism Spectrum Disorder (ASD) is the synaptic plasticity dysfunction, yet the molecular mechanism remains elusive. As a prototype, we investigate the postsynaptic signal transduction network in glutamatergic neurons and integrate single-cell nucleus transcriptomics data from the Prefrontal Cortex (PFC) to unveil the malfunction of translation control. METHODS We devise an innovative and highly dependable pipeline to transform our acquired signal transduction network into an mRNA Signaling-Regulatory Network (mSiReN) and analyze it at the RNA level. We employ Cell-Specific Network Inference via Integer Value Programming and Causal Reasoning (CS-NIVaCaR) to identify core modules and Cell-Specific Probabilistic Contextualization for mRNA Regulatory Networks (CS-ProComReN) to quantitatively reveal activated sub-pathways involving MAPK1, MKNK1, RPS6KA5, and MTOR across different cell types in ASD. RESULTS The results indicate that specific pivotal molecules, such as EIF4EBP1 and EIF4E, lacking Differential Expression (DE) characteristics and responsible for protein translation with long-term potentiation (LTP) or long-term depression (LTD), are dysregulated. We further uncover distinct activation patterns causally linked to the EIF4EBP1-EIF4E module in excitatory and inhibitory neurons. CONCLUSIONS Importantly, our work introduces a methodology for leveraging extensive transcriptomics data to parse the signal transduction network, transforming it into mSiReN, and mapping it back to the protein level. These algorithms can serve as potent tools in systems biology to analyze other omics and regulatory networks. Furthermore, the biomarkers within the activated sub-pathways, revealed by identifying convergent dysregulation, illuminate potential diagnostic and prognostic factors in ASD.
Collapse
Affiliation(s)
- Chao Kong
- School of Systems Science, Beijing Normal University, Beijing 100875, China;
| | - Zhitong Bing
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Lei Yang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Zigang Huang
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Wenxu Wang
- School of Systems Science, Beijing Normal University, Beijing 100875, China;
| | - Celso Grebogi
- Institute for Complex Systems and Mathematical Biology, King’s College, University of Aberdeen, Old Aberdeen AB24 3UE, UK
| |
Collapse
|
127
|
Dominguez G, Wu Y, Zhou J. Epigenetic Regulation and Neurodevelopmental Disorders: From MeCP2 to the TCF20/PHF14 Complex. Genes (Basel) 2024; 15:1653. [PMID: 39766920 PMCID: PMC11728296 DOI: 10.3390/genes15121653] [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: 12/01/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) affect approximately 15% of children and adolescents worldwide. This group of disorders is often polygenic with varying risk factors, with many associated genes converging on shared molecular pathways, including chromatin regulation and transcriptional control. Understanding how NDD-associated chromatin regulators and protein complexes orchestrate these regulatory pathways is crucial for elucidating NDD pathogenesis and developing targeted therapeutic strategies. Recently, the TCF20/PHF14 chromatin complex was identified in the mammalian brain, expanding the list of chromatin regulatory remodelers implicated in NDDs. This complex-which includes MeCP2, RAI1, TCF20, PHF14, and HMG20A-plays a vital role in epigenetic and transcriptional regulation. METHODS We review and summarize current research and clinical reports pertaining to the different components of the MeCP2-interacting TCF20/PHF14 complex. We examine the NDDs associated with the TCF20/PHF14 complex, explore the molecular and neuronal functions of its components, and discuss emerging therapeutic strategies targeting this complex to mitigate symptoms, with broader applicability to other NDDs. RESULTS Mutations in the genes encoding the components of the MeCP2-interacting TCF20/PHF14 complex have been linked to various NDDs, underscoring its critical contribution to brain development and NDD pathogenesis. CONCLUSIONS The MeCP2-interacting TCF20/PHF14 complex and its associated NDDs could serve as a model system to provide insight into the interplay between epigenetic regulation and NDD pathogenesis.
Collapse
Affiliation(s)
- Gaea Dominguez
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (G.D.); (Y.W.)
| | - Yongji Wu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (G.D.); (Y.W.)
| | - Jian Zhou
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (G.D.); (Y.W.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
128
|
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
|
129
|
Ma K, McDaniel K, Zhang D, Webb M, Qin L. Chemogenetic Inhibition of Prefrontal Cortex Ameliorates Autism-Like Social Deficits and Absence-Like Seizures in a Gene-Trap Ash1l Haploinsufficiency Mouse Model. Genes (Basel) 2024; 15:1619. [PMID: 39766886 PMCID: PMC11675260 DOI: 10.3390/genes15121619] [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: 11/26/2024] [Revised: 12/08/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND ASH1L (absent, small, or homeotic-like 1), a histone methyltransferase, has been identified as a high-risk gene for autism spectrum disorder (ASD). We previously showed that postnatal Ash1l severe deficiency in the prefrontal cortex (PFC) of male and female mice caused seizures. However, the synaptic mechanisms underlying autism-like social deficits and seizures need to be elucidated. OBJECTIVE The goal of this study is to characterize the behavioral deficits and reveal the synaptic mechanisms in an Ash1l haploinsufficiency mouse model using a targeted gene-trap knockout (gtKO) strategy. METHOD A series of behavioral tests were used to examine behavioral deficits. Electrophysiological and chemogenetic approaches were used to examine and manipulate the excitability of pyramidal neurons in the PFC of Ash1l+/GT mice. RESULTS Ash1l+/GT mice displayed social deficits, increased self-grooming, and cognitive impairments. Epileptiform discharges were found on electroencephalograms (EEGs) of Ash1l+/GT mice, indicating absence-like seizures. Ash1l haploinsufficiency increased the susceptibility for convulsive seizures when Ash1l+/GT mice were challenged by pentylenetetrazole (PTZ, a competitive GABAA receptor antagonist). Whole-cell patch-clamp recordings showed that Ash1l haploinsufficiency increased the excitability of pyramidal neurons in the PFC by altering intrinsic neuronal properties, enhancing glutamatergic synaptic transmission, and diminishing GABAergic synaptic inhibition. Chemogenetic inhibition of pyramidal neurons in the PFC of Ash1l+/GT mice ameliorated autism-like social deficits and abolished absence-like seizures. CONCLUSIONS We demonstrated that increased neural activity in the PFC contributed to the autism-like social deficits and absence-like seizures in Ash1l+/GT mice, which provides novel insights into the therapeutic strategies for patients with ASH1L-associated ASD and epilepsy.
Collapse
Affiliation(s)
- Kaijie Ma
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (K.M.)
| | - Kylee McDaniel
- Department of Biotechnology, Mount Marty University, Yankton, SD 57078, USA;
| | - Daoqi Zhang
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (K.M.)
| | - Maria Webb
- School of Health Sciences, University of South Dakota, Vermillion, SD 57069, USA
| | - Luye Qin
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (K.M.)
| |
Collapse
|
130
|
Huguet G, Renne T, Poulain C, Dubuc A, Kumar K, Kazem S, Engchuan W, Shanta O, Douard E, Proulx C, Jean-Louis M, Saci Z, Mollon J, Schultz LM, Knowles EEM, Cox SR, Porteous D, Davies G, Redmond P, Harris SE, Schumann G, Dumas G, Labbe A, Pausova Z, Paus T, Scherer SW, Sebat J, Almasy L, Glahn DC, Jacquemont S. Effects of gene dosage on cognitive ability: A function-based association study across brain and non-brain processes. CELL GENOMICS 2024; 4:100721. [PMID: 39667348 PMCID: PMC11701252 DOI: 10.1016/j.xgen.2024.100721] [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: 04/19/2024] [Revised: 07/30/2024] [Accepted: 11/13/2024] [Indexed: 12/14/2024]
Abstract
Copy-number variants (CNVs) that increase the risk for neurodevelopmental disorders also affect cognitive ability. However, such CNVs remain challenging to study due to their scarcity, limiting our understanding of gene-dosage-sensitive biological processes linked to cognitive ability. We performed a genome-wide association study (GWAS) in 258,292 individuals, which identified-for the first time-a duplication at 2q12.3 associated with higher cognitive performance. We developed a functional-burden analysis, which tested the association between cognition and CNVs disrupting 6,502 gene sets biologically defined across tissues, cell types, and ontologies. Among those, 864 gene sets were associated with cognition, and effect sizes of deletion and duplication were negatively correlated. The latter suggested that functions across all biological processes were sensitive to either deletions (e.g., subcortical regions, postsynaptic) or duplications (e.g., cerebral cortex, presynaptic). Associations between non-brain tissues and cognition were driven partly by constrained genes, which may shed light on medical comorbidities in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Guillaume Huguet
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada.
| | - Thomas Renne
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Cécile Poulain
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Alma Dubuc
- École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Kuldeep Kumar
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Sayeh Kazem
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Worrawat Engchuan
- The Hospital for Sick Children, Genetics and Genome Biology, Toronto, ON, Canada; The Hospital for Sick Children, The Centre for Applied Genomics, Toronto, ON, Canada
| | - Omar Shanta
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Elise Douard
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Catherine Proulx
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Martineau Jean-Louis
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Zohra Saci
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Josephine Mollon
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura M Schultz
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emma E M Knowles
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon R Cox
- Lothian Birth Cohorts, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - David Porteous
- Lothian Birth Cohorts, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh EH8 9JZ, UK; Medical Genetics Section, Centre for Genomic & Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK; Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Gail Davies
- Lothian Birth Cohorts, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Paul Redmond
- Lothian Birth Cohorts, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Sarah E Harris
- Lothian Birth Cohorts, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Gunter Schumann
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Guillaume Dumas
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Mila - Quebec Artificial Intelligence Institute, Montréal, QC, Canada
| | - Aurélie Labbe
- Département de Sciences de la Décision, HEC Montreal, Montreal, QC, Canada
| | - Zdenka Pausova
- Research Institute of the Hospital for Sick Children, Toronto, ON, Canada; Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada; ECOGENE-21, Chicoutimi, QC, Canada
| | - Tomas Paus
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Department of Psychiatry and Addictology, Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Stephen W Scherer
- The Hospital for Sick Children, Genetics and Genome Biology, Toronto, ON, Canada; The Hospital for Sick Children, The Centre for Applied Genomics, Toronto, ON, Canada; McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Jonathan Sebat
- University of California, San Diego, Department of Psychiatry, Department of Cellular & Molecular Medicine, Beyster Center of Psychiatric Genomics, San Diego, CA, USA
| | - Laura Almasy
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David C Glahn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Olin Neuropsychiatric Research Center, Institute of Living, Hartford Hospital, Hartford, CT, USA
| | - Sébastien Jacquemont
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Department of Pediatrics, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
131
|
Jung S, Caballero M, Kępińska A, Smout S, Munk-Olsen T, Robakis TK, Bergink V, Mahjani B. Genetic Architecture of Postpartum Psychosis: From Common to Rare Genetic Variation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.09.24318732. [PMID: 39711717 PMCID: PMC11661424 DOI: 10.1101/2024.12.09.24318732] [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: 12/24/2024]
Abstract
Postpartum psychosis is a severe psychiatric condition marked by the abrupt onset of psychosis, mania, or psychotic depression following childbirth. Despite evidence for a strong genetic basis, the roles of common and rare genetic variation remain poorly understood. Leveraging data from Swedish national registers and genomic data from the All of Us Research Program, we estimated family-based heritability at 55% and WGS-based heritability at 37%, with an overrepresentation on the X chromosome. Rare coding variant analysis identified DNMT1 and HMGCR as potential risk genes (q < 0.1). Analysis of 240,009 samples from All of Us demonstrated significant associations between these genes and multiple psychiatric disorders, supporting their biological relevance. Additionally, 17% of bipolar disorder, 21% of schizophrenia, and 16-25% of multiple autoimmune disorder risk genes overlapped with postpartum psychosis. These findings reveal unique genetic contributions and shared pathways, providing a foundation for understanding pathophysiology and advancing therapeutic strategies.
Collapse
Affiliation(s)
- Seulgi Jung
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madison Caballero
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adrianna Kępińska
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shelby Smout
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Trine Munk-Olsen
- Department of Clinical Research, Research Unit Children and Adolescent Psychiatry, University of Southern Denmark, Denmark
| | - Thalia K. Robakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Veerle Bergink
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Behrang Mahjani
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
132
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: implications for brain development and autism. Transl Psychiatry 2024; 14:482. [PMID: 39632793 PMCID: PMC11618798 DOI: 10.1038/s41398-024-03179-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood-stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNA-seq data obtained from E12.5 fetal mouse brains 3 hours after VPA administration to the pregnant dam revealed that VPA rapidly and significantly increased or decreased the expression of approximately 7,300 genes. No significant sex differences in VPA-induced gene expression were observed. Expression of 399 autism risk genes was significantly altered by VPA as was expression of 258 genes that have been reported to modulate fetal brain development but are not otherwise linked to autism. Expression of genes associated with intracellular signaling pathways, neurogenesis, and excitation-inhibition balance as well as synaptogenesis, neuronal fate determination, axon and dendritic development, neuroinflammation, circadian rhythms, and epigenetic modulation of gene expression was dysregulated by VPA. Notably, at least 40 genes that are known to regulate embryonic neurogenesis were dysregulated by VPA. The goal of this study was to identify mouse genes that are: (a) significantly up- or downregulated by VPA in the fetal brain and (b) associated with autism and/or known to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity and, consequently behavior, in the adult. The genes meeting these criteria provide potential targets for future hypothesis-driven studies to elucidate the proximal causes of errors in brain connectivity underlying neurodevelopmental disorders such as autism.
Collapse
Affiliation(s)
- Susan G Dorsey
- Department of Pain and Translational Symptom Science University of Maryland School of Nursing, Baltimore, MD, 21201, USA
| | - Evelina Mocci
- Department of Pain and Translational Symptom Science University of Maryland School of Nursing, Baltimore, MD, 21201, USA
- Institute for Genome Sciences University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Malcolm V Lane
- Translational Toxicology/Department of Epidemiology and Public Health University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Bruce K Krueger
- Departments of Physiology and Psychiatry University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
133
|
Kshetri R, Beavers JO, Hyde R, Ewa R, Schwertman A, Porcayo S, Richardson BD. Behavioral decline in Shank3 Δex4-22 mice during early adulthood parallels cerebellar granule cell glutamatergic synaptic changes. Mol Autism 2024; 15:52. [PMID: 39633421 PMCID: PMC11616285 DOI: 10.1186/s13229-024-00628-y] [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: 08/09/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND SHANK3, a gene encoding a synaptic scaffolding protein, is implicated in autism spectrum disorder (ASD) and is disrupted in Phelan-McDermid syndrome (PMS). Despite evidence of regression or worsening of ASD-like symptoms in individuals with PMS, the underlying mechanisms remain unclear. Although Shank3 is highly expressed in the cerebellar cortical granule cells, its role in cerebellar function and contribution to behavioral deficits in ASD models are unknown. This study investigates behavioral changes and cerebellar synaptic alterations in Shank3Δex4-22 mice at two developmental stages. METHODS Shank3Δex4-22 wildtype, heterozygous, and homozygous knockout mice lacking exons 4-22 (all functional isoforms) were subjected to a behavioral battery in both juvenile (5-7 weeks old) and adult (3-5 months old) mouse cohorts of both sexes. Immunostaining was used to show the expression of Shank3 in the cerebellar cortex. Spontaneous excitatory postsynaptic currents (sEPSCs) from cerebellar granule cells (CGCs) were recorded by whole-cell patch-clamp electrophysiology. RESULTS Deletion of Shank3 caused deficits in motor function, heightened anxiety, and repetitive behaviors. These genotype-dependent behavioral alterations were more prominent in adult mice than in juveniles. Reduced social preference was only identified in adult Shank3Δex4-22 knockout male mice, while self-grooming was uniquely elevated in males across both age groups. Heterozygous mice showed little to no changes in behavioral phenotypes in most behavioral tests. Immunofluorescence staining indicated the presence of Shank3 predominantly in the dendrite-containing rosette-like structures in CGCs, colocalizing with presynaptic markers of glutamatergic mossy fiber. Electrophysiological findings identified a parallel relationship between the age-related exacerbation of behavioral impairments and the enhancement of sEPSC amplitude in CGCs. LIMITATIONS Other behavioral tests of muscle strength (grip strength test), memory (Barnes/water maze), and communication (ultrasonic vocalization), were not performed. Further study is necessary to elucidate how Shank3 modulates synaptic function at the mossy fiber-granule cell synapse in the cerebellum and whether these changes shape the behavioral phenotype. CONCLUSIONS Our findings reveal an age-related exacerbation of behavioral impairments in Shank3Δex4-22 mutant mice. These results suggest that Shank3 may alter the function of glutamatergic receptors at the mossy fiber-cerebellar granule cell synapse as a potential mechanism causing cerebellar disruption in ASD.
Collapse
Affiliation(s)
- Rajaram Kshetri
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - James O Beavers
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Romana Hyde
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Roseline Ewa
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Amber Schwertman
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Sarahi Porcayo
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Ben D Richardson
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA.
| |
Collapse
|
134
|
Gogate A, Kaur K, Khalil R, Bashtawi M, Morris MA, Goodspeed K, Evans P, Chahrour MH. The genetic landscape of autism spectrum disorder in an ancestrally diverse cohort. NPJ Genom Med 2024; 9:62. [PMID: 39632905 PMCID: PMC11618689 DOI: 10.1038/s41525-024-00444-6] [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: 05/15/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Autism spectrum disorder (ASD) comprises neurodevelopmental disorders with wide variability in genetic causes and phenotypes, making it challenging to pinpoint causal genes. We performed whole exome sequencing on a modest, ancestrally diverse cohort of 195 families, including 754 individuals (222 with ASD), and identified 38,834 novel private variants. In 68 individuals with ASD (~30%), we identified 92 potentially pathogenic variants in 73 known genes, including BCORL1, CDKL5, CHAMP1, KAT6A, MECP2, and SETD1B. Additionally, we identified 158 potentially pathogenic variants in 120 candidate genes, including DLG3, GABRQ, KALRN, KCTD16, and SLC8A3. We also found 34 copy number variants in 31 individuals overlapping known ASD loci. Our work expands the catalog of ASD genetics by identifying hundreds of variants across diverse ancestral backgrounds, highlighting convergence on nervous system development and signal transduction. These findings provide insights into the genetic underpinnings of ASD and inform molecular diagnosis and potential therapeutic targets.
Collapse
Affiliation(s)
- Ashlesha Gogate
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kiran Kaur
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Raida Khalil
- Department of Biotechnology and Genetic Engineering, Faculty of Science, University of Philadelphia, Amman, Jordan
| | - Mahmoud Bashtawi
- Department of Psychiatry, Jordan University of Science and Technology, King Abdullah University Hospital, Ramtha, Jordan
| | - Mary Ann Morris
- UT Southwestern and Children's Health Center for Autism Care, Children's Medical Center Dallas, Dallas, TX, 75247, USA
| | - Kimberly Goodspeed
- UT Southwestern and Children's Health Center for Autism Care, Children's Medical Center Dallas, Dallas, TX, 75247, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Patricia Evans
- UT Southwestern and Children's Health Center for Autism Care, Children's Medical Center Dallas, Dallas, TX, 75247, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Maria H Chahrour
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
135
|
Jhanji M, Ward JA, Leung CS, Krall CL, Ritchie FD, Guevara A, Vestergaard K, Yoon B, Amin K, Berto S, Liu J, Lizarraga SB. Dynamic Regulation OF The Chromatin Environment By Ash1L Modulates Human Neuronal Structure And Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.625500. [PMID: 39677608 PMCID: PMC11642754 DOI: 10.1101/2024.12.02.625500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Precise regulation of the chromatin environment through post-translational histone modification modulates transcription and controls brain development. Not surprisingly, mutations in a large number of histone-modifying enzymes underlie complex brain disorders. In particular, the histone methyltransferase ASH1L modifies histone marks linked to transcriptional activation and has been implicated in multiple neuropsychiatric disorders. However, the mechanisms underlying the pathobiology of ASH1L-asociated disease remain underexplored. We generated human isogenic stem cells with a mutation in ASH1L's catalytic domain. We find that ASH1L dysfunction results in reduced neurite outgrowth, which correlates with alterations in the chromatin profile of activating and repressive histone marks, as well as the dysregulation of gene programs important for neuronal structure and function implicated in neuropsychiatric disease. We also identified a novel regulatory node implicating both the SP and Krüppel -like families of transcription factors and ASH1L relevant to human neuronal development. Finally, we rescue cellular defects linked to ASH1L dysfunction by leveraging two independent epigenetic mechanisms that promote transcriptional activation. In summary, we identified an ASH1L-driven epigenetic and transcriptional axis essential for human brain development and complex brain disorders that provide insights into future therapeutic strategies for ASH1L-related disorders.
Collapse
|
136
|
Séjourné G, Eroglu C. Astrocyte-neuron crosstalk in neurodevelopmental disorders. Curr Opin Neurobiol 2024; 89:102925. [PMID: 39357429 DOI: 10.1016/j.conb.2024.102925] [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: 06/25/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 10/04/2024]
Abstract
A fundamental feature shared across neurodevelopmental disorders (NDDs) is the disruption of synaptic circuit formation and homeostasis. During early life, non-neuronal cells called astrocytes tightly regulate the establishment of circuits by controlling formation, remodeling, stabilization, and maturation of synapses. Concurrently, astrocytes mature to meet the evolving needs of the developing brain. Bidirectional astrocyte-neuron communication synchronizes astrocyte maturation with synapse development. An emerging body of evidence supports the hypothesis that in NDDs, deficits in astrocyte-neuron communication underlie errors in synaptic circuit development. Here we will review and discuss these findings, with the aim of inspiring future research and guiding translational studies.
Collapse
Affiliation(s)
- Gabrielle Séjourné
- The Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| | - Cagla Eroglu
- The Department of Cell Biology, Duke University Medical Center, Durham, NC, USA; The Department of Neurobiology, Duke University Medical Center, Durham, NC, USA; Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
137
|
Senko K, Saddoris KL, Baus E, Soe K, Vaughn SE. Catatonia responsive to corticosteroids in a patient with an SCN2A variant. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2024; 196:e32101. [PMID: 39056510 DOI: 10.1002/ajmg.c.32101] [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: 05/31/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Variants in SCN2A are a known risk factor for developing autism spectrum disorder (ASD). Catatonia is a complex neuropsychiatric syndrome, which occurs at a higher rate in individuals with ASD. Catatonia has also been associated with COVID-19 infection, though the majority of these cases are associated with increased serum inflammatory markers. We present a case of a 15-year-old female with ASD and corticosteroid responsive stuporous catatonia to explore the relationship between SCN2A variants, ASD, COVID-19 exposure, and treatment refractory catatonia. Despite a lack of significantly elevated serum or CSF inflammatory markers, this patient showed significant improvement following initiation of corticosteroid therapy. This case presents a novel approach to the work-up and treatment of catatonia in individuals with SCN2A variants independent of elevated inflammatory markers.
Collapse
Affiliation(s)
- Kimberly Senko
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Child & Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kelsey L Saddoris
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Child & Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ella Baus
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Child & Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Katherine Soe
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Child & Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Samuel E Vaughn
- Division of Child & Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
138
|
Chen G, Xu M, Chen Z, Yang F. Clinical applications of small-molecule GABA AR modulators for neurological disorders. Bioorg Chem 2024; 153:107983. [PMID: 39581171 DOI: 10.1016/j.bioorg.2024.107983] [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: 10/02/2024] [Revised: 10/31/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024]
Abstract
Gamma-aminobutyric acid type A receptor (GABAAR) modulators are crucial in treating neurological and psychiatric disorders, including epilepsy, anxiety, insomnia, and depression. This review examines the synthetic approaches and clinical applications of representative small-molecule GABAAR modulators. Benzodiazepines, such as Diazepam, are well-known positive allosteric modulators (PAMs) that enhance GABAAR function, providing therapeutic effects but also associated with side effects like sedation and dependence. Non-benzodiazepine modulators, including Z-drugs like Zolpidem and Zaleplon, offer improved selectivity for the α1 subunit of GABAAR, reducing some of these side effects. Neurosteroids such as allopregnanolone and its synthetic analogs, including Brexanolone, have emerged as potent GABAAR modulators with applications in conditions like postpartum depression and refractory epilepsy. Advances in molecular biology and pharmacology have facilitated the development of isoform-specific modulators, potentially reducing off-target effects and enhancing therapeutic profiles. Additionally, combining GABAAR modulators with other therapeutic agents has shown promise in enhancing efficacy and minimizing side effects. This review highlights the design strategies, pharmacodynamics, clinical efficacy, and safety profiles of these compounds, emphasizing the opportunities for developing novel GABAAR modulators with improved therapeutic outcomes.
Collapse
Affiliation(s)
- Guangyong Chen
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meiling Xu
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhuo Chen
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Fuwei Yang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
139
|
Miyake N, Tsurusaki Y, Fukai R, Kushima I, Okamoto N, Ohashi K, Nakamura K, Hashimoto R, Hiraki Y, Son S, Kato M, Sakai Y, Osaka H, Deguchi K, Matsuishi T, Takeshita S, Fattal-Valevski A, Ekhilevitch N, Tohyama J, Yap P, Keng WT, Kobayashi H, Takubo K, Okada T, Saitoh S, Yasuda Y, Murai T, Nakamura K, Ohga S, Matsumoto A, Inoue K, Saikusa T, Hershkovitz T, Kobayashi Y, Morikawa M, Ito A, Hara T, Uno Y, Seiwa C, Ishizuka K, Shirahata E, Fujita A, Koshimizu E, Miyatake S, Takata A, Mizuguchi T, Ozaki N, Matsumoto N. Molecular diagnosis of 405 individuals with autism spectrum disorder. Eur J Hum Genet 2024; 32:1551-1558. [PMID: 36973392 PMCID: PMC11606949 DOI: 10.1038/s41431-023-01335-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/08/2023] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Autism spectrum disorder (ASD) is caused by combined genetic and environmental factors. Genetic heritability in ASD is estimated as 60-90%, and genetic investigations have revealed many monogenic factors. We analyzed 405 patients with ASD using family-based exome sequencing to detect disease-causing single-nucleotide variants (SNVs), small insertions and deletions (indels), and copy number variations (CNVs) for molecular diagnoses. All candidate variants were validated by Sanger sequencing or quantitative polymerase chain reaction and were evaluated using the American College of Medical Genetics and Genomics/Association for Molecular Pathology guidelines for molecular diagnosis. We identified 55 disease-causing SNVs/indels in 53 affected individuals and 13 disease-causing CNVs in 13 affected individuals, achieving a molecular diagnosis in 66 of 405 affected individuals (16.3%). Among the 55 disease-causing SNVs/indels, 51 occurred de novo, 2 were compound heterozygous (in one patient), and 2 were X-linked hemizygous variants inherited from unaffected mothers. The molecular diagnosis rate in females was significantly higher than that in males. We analyzed affected sibling cases of 24 quads and 2 quintets, but only one pair of siblings shared an identical pathogenic variant. Notably, there was a higher molecular diagnostic rate in simplex cases than in multiplex families. Our simulation indicated that the diagnostic yield is increasing by 0.63% (range 0-2.5%) per year. Based on our simple simulation, diagnostic yield is improving over time. Thus, periodical reevaluation of ES data should be strongly encouraged in undiagnosed ASD patients.
Collapse
Affiliation(s)
- Noriko Miyake
- Department of Human Genetics, National Center for Global Health and Medicine, Tokyo, Japan.
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Yoshinori Tsurusaki
- Faculty of Nutritional Science, Sagami Women's University, Sagamihara, Japan
| | - Ryoko Fukai
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Kei Ohashi
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kazuhiko Nakamura
- Department of Neuropsychiatry, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ryota Hashimoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Yoko Hiraki
- Hiroshima Municipal Center for Child Health and Development, Hiroshima, Japan
| | - Shuraku Son
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | | | - Toyojiro Matsuishi
- Departments of Pediatrics and Child Health, Kurume University School of Medicine, Kurume, Japan
- Department of Pediatrics, St. Mary's Hospital, Kurume, Japan
| | - Saoko Takeshita
- Department of Pediatrics, Yokohama City University Medical Center, Yokohama, Japan
| | - Aviva Fattal-Valevski
- Pediatric Neurology Institute, Dana-Dwek Children's Hospital, Tel Aviv Medical Center & Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nina Ekhilevitch
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Jun Tohyama
- Department of Child Neurology, National Hospital Organization Nishiniigata Chuo Hospital, Niigata, Japan
| | - Patrick Yap
- Genetic Health Service New Zealand, Auckland, New Zealand
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Wee Teik Keng
- Genetic Department, Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Hiroshi Kobayashi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Keiyo Takubo
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takashi Okada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Developmental Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuka Yasuda
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Toshiya Murai
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuyuki Nakamura
- Department of Pediatrics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ayumi Matsumoto
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Ken Inoue
- Deguchi Pediatric Clinic, Omura, Japan
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Tomoko Saikusa
- Departments of Pediatrics and Child Health, Kurume University School of Medicine, Kurume, Japan
- Department of Pediatrics, St. Mary's Hospital, Kurume, Japan
| | - Tova Hershkovitz
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Yu Kobayashi
- Department of Child Neurology, National Hospital Organization Nishiniigata Chuo Hospital, Niigata, Japan
| | - Mako Morikawa
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Aiko Ito
- Department of Pediatrics, Yamagata Prefectural Rehabilitation Center for Children with Disabilities, Yamagata, Japan
| | | | - Yota Uno
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chizuru Seiwa
- Department of Pediatrics, Yamagata Prefectural Rehabilitation Center for Children with Disabilities, Yamagata, Japan
| | - Kanako Ishizuka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Emi Shirahata
- Department of Pediatrics, Yamagata Prefectural Rehabilitation Center for Children with Disabilities, Yamagata, Japan
| | - Atsushi Fujita
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Eriko Koshimizu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Clinical Genetics, Yokohama City University Hospital, Yokohama, Japan
| | - Atsushi Takata
- Laboratory for Molecular Pathology of Psychiatric Disorders, RIKEN Center for Brain Science, Wako, Japan
| | - Takeshi Mizuguchi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| |
Collapse
|
140
|
Rusina E, Simonti M, Duprat F, Cestèle S, Mantegazza M. Voltage-gated sodium channels in genetic epilepsy: up and down of excitability. J Neurochem 2024; 168:3872-3890. [PMID: 37654020 PMCID: PMC11591406 DOI: 10.1111/jnc.15947] [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: 07/16/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
The past two decades have witnessed a wide range of studies investigating genetic variants of voltage-gated sodium (NaV) channels, which are involved in a broad spectrum of diseases, including several types of epilepsy. We have reviewed here phenotypes and pathological mechanisms of genetic epilepsies caused by variants in NaV α and β subunits, as well as of some relevant interacting proteins (FGF12/FHF1, PRRT2, and Ankyrin-G). Notably, variants of all these genes can induce either gain- or loss-of-function of NaV leading to either neuronal hyperexcitability or hypoexcitability. We present the results of functional studies obtained with different experimental models, highlighting that they should be interpreted considering the features of the experimental system used. These systems are models, but they have allowed us to better understand pathophysiological issues, ameliorate diagnostics, orientate genetic counseling, and select/develop therapies within a precision medicine framework. These studies have also allowed us to gain insights into the physiological roles of different NaV channels and of the cells that express them. Overall, our review shows the progress that has been made, but also the need for further studies on aspects that have not yet been clarified. Finally, we conclude by highlighting some significant themes of general interest that can be gleaned from the results of the work of the last two decades.
Collapse
Affiliation(s)
- Evgeniia Rusina
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
| | - Martina Simonti
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
| | - Fabrice Duprat
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
- InsermValbonne‐Sophia AntipolisFrance
| | - Sandrine Cestèle
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
| | - Massimo Mantegazza
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
- InsermValbonne‐Sophia AntipolisFrance
| |
Collapse
|
141
|
Jaudon F, Cingolani LA. Unlocking mechanosensitivity: integrins in neural adaptation. Trends Cell Biol 2024; 34:1029-1043. [PMID: 38514304 DOI: 10.1016/j.tcb.2024.02.011] [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: 01/07/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Mechanosensitivity extends beyond sensory cells to encompass most neurons in the brain. Here, we explore recent research on the role of integrins, a diverse family of adhesion molecules, as crucial biomechanical sensors translating mechanical forces into biochemical and electrical signals in the brain. The varied biomechanical properties of neuronal integrins, including their force-dependent conformational states and ligand interactions, dictate their specific functions. We discuss new findings on how integrins regulate filopodia and dendritic spines, shedding light on their contributions to synaptic plasticity, and explore recent discoveries on how they engage with metabotropic receptors and ion channels, highlighting their direct participation in electromechanical transduction. Finally, to facilitate a deeper understanding of these developments, we present molecular and biophysical models of mechanotransduction.
Collapse
Affiliation(s)
- Fanny Jaudon
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Lorenzo A Cingolani
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; Center for Synaptic Neuroscience and Technology (NSYN), Fondazione Istituto Italiano di Tecnologia (IIT), 16132 Genoa, Italy.
| |
Collapse
|
142
|
Aldridge AI, West AE. Epigenetics and the timing of neuronal differentiation. Curr Opin Neurobiol 2024; 89:102915. [PMID: 39277975 PMCID: PMC11611672 DOI: 10.1016/j.conb.2024.102915] [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: 06/16/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/17/2024]
Abstract
Epigenetic regulation of the genome is required for cell-type differentiation during organismal development and is especially important to generate the panoply of specialized cell types that comprise the brain. Here, we review how progressive changes in the chromatin landscape, both in neural progenitors and in postmitotic neurons, orchestrate the timing of gene expression programs that underlie first neurogenesis and then functional neuronal maturation. We discuss how disease-associated mutations in chromatin regulators can change brain composition by impairing the timing of neurogenesis. Further, we highlight studies that are beginning to show how chromatin modifications are integrated at the level of chromatin architecture to coordinate changing transcriptional programs across developmental including in postmitotic neurons.
Collapse
Affiliation(s)
- Andrew I Aldridge
- Duke University School of Medicine, Department of Neurobiology, Durham, NC 27710, USA
| | - Anne E West
- Duke University School of Medicine, Department of Neurobiology, Durham, NC 27710, USA.
| |
Collapse
|
143
|
Zhao S, Li M, Chen WJ, Rennie BJ, Hsiao YY, Guan Y. Parental experiences and perspectives of healthcare providers' genetic testing recommendations for their children diagnosed with autism spectrum disorder in the United States. Autism Res 2024; 17:2471-2486. [PMID: 39533966 DOI: 10.1002/aur.3262] [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: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Although genetic testing is recommended for all children diagnosed with autism spectrum disorder (ASD) in the United States, it remains unclear how many parents have received and followed these recommendations. This study aimed to assess parental experiences and perspectives of healthcare providers' genetic testing recommendations. A total of 1043 parents of children with ASD from Simons Foundation Powering Autism Research for Knowledge completed an online survey. Only about one-third of the parents (34.2%) reported that their children's providers had recommended genetic testing. Factors associated with whether parents received these recommendations included the type of healthcare providers diagnosing ASD, parents' knowledge of genetic testing, the age of their children's diagnosis, and the presence of certain co-occurring medical conditions in their children. Our study also revealed that most parents (76.9%) who received recommendations had pursued genetic testing for their children. Adherence to the recommendations was associated with the characteristics of the parents (i.e., age and employment status) and children (i.e., gender), and their trust in the information from providers. The findings highlight the critical role of healthcare providers in facilitating genetic testing among families of children with ASD. Healthcare providers are recommended to increase their competencies and practice in providing genetic testing recommendations for ASD, assist parents in navigating testing challenges, and support parents through the testing process.
Collapse
Affiliation(s)
- Shixi Zhao
- College of Population Health, University of New Mexico, Albuquerque, New Mexico, USA
| | - Ming Li
- Department of Health Sciences, Towson University, Towson, Maryland, USA
| | - Wei-Ju Chen
- Department of Psychology, The University of Texas Permian Basin, Odessa, Texas, USA
| | - Brandon J Rennie
- Department of Pediatrics, University of New Mexico, Albuquerque, New Mexico, USA
| | - Yu-Yu Hsiao
- Department of Individual, Family, & Community Education, University of New Mexico, Albuquerque, New Mexico, USA
| | - Yue Guan
- Department of Behavioral, Social, and Health Education Sciences, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
144
|
Haddad S, Hessenberger M, Ablinger C, Eibl C, Stanika R, Campiglio M, Obermair GJ. Autism-Linked Mutations in α 2δ-1 and α 2δ-3 Reduce Protein Membrane Expression but Affect Neither Calcium Channels nor Trans-Synaptic Signaling. Pharmaceuticals (Basel) 2024; 17:1608. [PMID: 39770450 PMCID: PMC11677996 DOI: 10.3390/ph17121608] [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: 10/17/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND α2δ proteins regulate membrane trafficking and biophysical properties of voltage-gated calcium channels. Moreover, they modulate axonal wiring, synapse formation, and trans-synaptic signaling. Several rare missense variants in CACNA2D1 (coding for α2δ-1) and CACNA2D3 (coding for α2δ-3) genes were identified in patients with autism spectrum disorder (ASD). However, the pathogenicity of these variants is not known, and the molecular mechanism by which α2δ proteins may contribute to the pathophysiology of autism is, as of today, not understood. Therefore, in this study we functionally characterized two heterozygous missense variants in α2δ-1 (p.R351T) and α2δ-3 (p.A275T), previously identified in patients with ASD. METHODS Electrophysiological recordings in transfected tsA201 cells were used to study specific channel-dependent functions of mutated α2δ proteins. Membrane expression, presynaptic targeting, and trans-synaptic signaling of mutated α2δ proteins were studied upon expression in murine cultured hippocampal neurons. RESULTS Homologous expression of both mutated α2δ proteins revealed a strongly reduced membrane expression and synaptic localization compared to the corresponding wild type α2δ proteins. Moreover, the A275T mutation in α2δ-3 resulted in an altered glycosylation pattern upon heterologous expression. However, neither of the mutations compromised the biophysical properties of postsynaptic L-type (CaV1.2 and CaV1.3) and presynaptic P/Q-type (CaV2.1) channels when co-expressed in tsA201 cells. Furthermore, presynaptic expression of p.R351T in the α2δ-1 splice variant lacking exon 23 did not affect trans-synaptic signaling to postsynaptic GABAA receptors. CONCLUSIONS Our data provide evidence that the pathophysiological mechanisms of ASD-causing mutations of α2δ proteins may not involve their classical channel-dependent and trans-synaptic functions. Alternatively, these mutations may induce subtle changes in synapse formation or neuronal network function, highlighting the need for future α2δ protein-linked disease models.
Collapse
Affiliation(s)
- Sabrin Haddad
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Manuel Hessenberger
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| | - Cornelia Ablinger
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Clarissa Eibl
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| | - Ruslan Stanika
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| | - Marta Campiglio
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Gerald J. Obermair
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| |
Collapse
|
145
|
Yen A, Sarafinovska S, Chen X, Skinner DD, Leti F, Crosby M, Hoisington-Lopez J, Wu Y, Chen J, Li ZA, Noguchi KK, Mitra RD, Dougherty JD. MYT1L deficiency impairs excitatory neuron trajectory during cortical development. Nat Commun 2024; 15:10308. [PMID: 39604385 PMCID: PMC11603064 DOI: 10.1038/s41467-024-54371-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Mutations reducing the function of MYT1L, a neuron-specific transcription factor, are associated with a syndromic neurodevelopmental disorder. MYT1L is used as a pro-neural factor in fibroblast-to-neuron transdifferentiation and is hypothesized to influence neuronal specification and maturation, but it is not clear which neuron types are most impacted by MYT1L loss. In this study, we profile 412,132 nuclei from the forebrains of wild-type and MYT1L-deficient mice at three developmental stages: E14 at the peak of neurogenesis, P1 when cortical neurons have been born, and P21 when neurons are maturing, to examine the role of MYT1L levels on neuronal development. MYT1L deficiency disrupts cortical neuron proportions and gene expression, primarily affecting neuronal maturation programs. Effects are mostly cell autonomous and persistent through development. While MYT1L can both activate and repress gene expression, the repressive effects are most sensitive to haploinsufficiency, likely mediating MYT1L syndrome. These findings illuminate MYT1L's role in orchestrating gene expression during neuronal development, providing insights into the molecular underpinnings of MYT1L syndrome.
Collapse
Affiliation(s)
- Allen Yen
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Simona Sarafinovska
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Xuhua Chen
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | | | | | - MariaLynn Crosby
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO, USA
- DNA Sequencing and Innovation Lab, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jessica Hoisington-Lopez
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO, USA
- DNA Sequencing and Innovation Lab, Washington University School of Medicine, Saint Louis, MO, USA
| | - Yizhe Wu
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jiayang Chen
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Zipeng A Li
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kevin K Noguchi
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Robi D Mitra
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA.
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
146
|
Haetzel LM, Iafrati J, Cording KR, Farhan M, Noveir SD, Rumbaugh G, Bateup HS. Haploinsufficiency of Syngap1 in Striatal Indirect Pathway Neurons Alters Motor and Goal-Directed Behaviors in Mice. J Neurosci 2024; 44:e1264232024. [PMID: 39358043 PMCID: PMC11604145 DOI: 10.1523/jneurosci.1264-23.2024] [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: 06/30/2023] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
SYNGAP1 is a high-confidence autism spectrum disorder (ASD) risk gene, and mutations in SYNGAP1 lead to a neurodevelopmental disorder (NDD) that presents with epilepsy, ASD, motor developmental delay, and intellectual disability. SYNGAP1 codes for Ras/Rap GTP-ase activating protein SynGAP (SynGAP). SynGAP is located in the postsynaptic density of glutamatergic synapses and regulates glutamate receptor trafficking in an activity-dependent manner. In addition to forebrain glutamatergic neurons, Syngap1 is highly expressed in the striatum, although the functions of SynGAP in the striatum have not been extensively studied. Here we show that Syngap1 is expressed in both direct and indirect pathway striatal projection neurons (dSPNs and iSPNs) in mice of both sexes. In a mouse model of Syngap1 haploinsufficiency, dendritic spine density, morphology, and intrinsic excitability are altered primarily in iSPNs, but not dSPNs. At the behavioral level, SynGAP reduction alters striatal-dependent motor learning and goal-directed behavior. Several behavioral phenotypes are reproduced by iSPN-specific Syngap1 reduction and, in turn, prevented by iSPN-specific Syngap1 rescue. These results establish the importance of SynGAP to striatal neuron function and pinpoint the indirect pathway as a key circuit in the neurobiology of SYNGAP1-related NDD.
Collapse
Affiliation(s)
- Laura M Haetzel
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Jillian Iafrati
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Katherine R Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
- Neuroscience, University of California, Berkeley, California 94720
| | - Mahmoud Farhan
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Sasan D Noveir
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Gavin Rumbaugh
- Departments of Neuroscience and Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458
- Skaggs Graduate School of Chemical and Biological Sciences, Jupiter, Florida 33458
| | - Helen S Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
- Neuroscience, University of California, Berkeley, California 94720
- Weill Neurohub Investigator, University of California, Berkeley, California 94720
| |
Collapse
|
147
|
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
|
148
|
Iwanicki T, Iwanicka J, Balcerzyk-Matić A, Jarosz A, Nowak T, Emich-Widera E, Kazek B, Kapinos-Gorczyca A, Kapinos M, Gawron K, Auguściak-Duma A, Likus W, Niemiec P. Association of CHD8 Gene Polymorphic Variants with the Clinical Phenotype of Autism Spectrum Disorder. J Clin Med 2024; 13:7019. [PMID: 39685474 DOI: 10.3390/jcm13237019] [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: 09/24/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Background: The CHD8 gene encodes chromodomain helicase DNA-binding protein 8 (CHD8), which is a transcriptional regulator involved in neuron development, myelination, and synaptogenesis. Some CHD8 gene mutations lead to neurodevelopmental syndromes with core symptoms of autism. The aim of this study was to perform an analysis of the family-based association of CHD8 gene polymorphisms with the occurrence and clinical phenotype of autism spectrum disorder (ASD). Methods: We analyzed 210 Caucasian children with ASD and their biological parents. The genotyping of specified polymorphisms, i.e., rs7148741, rs35057134, and rs10467770, was performed using TaqMan-PCR and compared with specific symptoms of ASD. Results: The G allele (rs7148741) was associated with muscle hypotonia as compared with the AA homozygotes. AA homozygosity (rs35057134) predisposed an individual to the use of an incubator, heart rate fluctuations, and the necessity of hospitalization. Moreover, the alleles and genotypes of this polymorphism were characterized by different Apgar scores and distributions. Additionally, CC homozygotes of rs10467770 were more often predisposed to the use of an incubator and hospitalization relative to T allele carriers. The average Apgar score was higher in TT homozygotes. Conclusions: Polymorphisms of the CHD8 gene may determine specific clinical phenotypes of ASD.
Collapse
Affiliation(s)
- Tomasz Iwanicki
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia in Katowice, Medykow Street 18, 40-752 Katowice, Poland
| | - Joanna Iwanicka
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia in Katowice, Medykow Street 18, 40-752 Katowice, Poland
| | - Anna Balcerzyk-Matić
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia in Katowice, Medykow Street 18, 40-752 Katowice, Poland
| | - Alicja Jarosz
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia in Katowice, Medykow Street 18, 40-752 Katowice, Poland
| | - Tomasz Nowak
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia in Katowice, Medykow Street 18, 40-752 Katowice, Poland
| | - Ewa Emich-Widera
- Department of Pediatric Neurology, Faculty of Medical Science in Katowice, Medical University of Silesia in Katowice, Medykow Street 16, 40-752 Katowice, Poland
| | - Beata Kazek
- Child Development Support Center Persevere, Kępowa Street 56, 40-583 Katowice, Poland
| | | | - Maciej Kapinos
- CZP Feniks, Daily Ward for Children and Adolescents, Młyńska Street 8, 44-100 Gliwice, Poland
| | - Katarzyna Gawron
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Medykow Street 18, 40-752 Katowice, Poland
| | - Aleksandra Auguściak-Duma
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Medykow Street 18, 40-752 Katowice, Poland
| | - Wirginia Likus
- Department of Anatomy, Faculty of Health Sciences in Katowice, Medical University of Silesia in Katowice, Medykow Street 18, 40-752 Katowice, Poland
| | - Paweł Niemiec
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia in Katowice, Medykow Street 18, 40-752 Katowice, Poland
| |
Collapse
|
149
|
Fan Y, Chen J, Fan Z, Chirinos J, Stein JL, Sullivan PF, Wang R, Nadig A, Zhang DY, Huang S, Jiang Z, Guan PY, Qian X, Li T, Li H, Sun Z, Ritchie MD, O’Brien J, Witschey W, Rader DJ, Li T, Zhu H, Zhao B. Mapping rare protein-coding variants on multi-organ imaging traits. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.16.24317443. [PMID: 39606337 PMCID: PMC11601754 DOI: 10.1101/2024.11.16.24317443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Human organ structure and function are important endophenotypes for clinical outcomes. Genome-wide association studies (GWAS) have identified numerous common variants associated with phenotypes derived from magnetic resonance imaging (MRI) of the brain and body. However, the role of rare protein-coding variations affecting organ size and function is largely unknown. Here we present an exome-wide association study that evaluates 596 multi-organ MRI traits across over 50,000 individuals from the UK Biobank. We identified 107 variant-level associations and 224 gene-based burden associations (67 unique gene-trait pairs) across all MRI modalities, including PTEN with total brain volume, TTN with regional peak circumferential strain in the heart left ventricle, and TNFRSF13B with spleen volume. The singleton burden model and AlphaMissense annotations contributed 8 unique gene-trait pairs including the association between an approved drug target gene of KCNA5 and brain functional activity. The identified rare coding signals elucidate some shared genetic regulation across organs, prioritize previously identified GWAS loci, and are enriched for drug targets. Overall, we demonstrate how rare variants enhance our understanding of genetic effects on human organ morphology and function and their connections to complex diseases.
Collapse
Affiliation(s)
- Yijun Fan
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jie Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julio Chirinos
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jason L. Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rujin Wang
- Regeneron Genetics Center, 777 Old Saw Mill River Rd., Tarrytown, NY, 10591, USA
| | - Ajay Nadig
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David Y. Zhang
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuai Huang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zhiwen Jiang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peter Yi Guan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xinjie Qian
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ting Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Haoyue Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zehui Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Joan O’Brien
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Medicine Center for Ophthalmic Genetics in Complex Diseases, Philadelphia, PA 19104, USA
| | - Walter Witschey
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel J. Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxin Zhao
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Population Aging Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Center for Eye-Brain Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
150
|
Teter OM, McQuade A, Hagan V, Liang W, Dräger NM, Sattler SM, Holmes BB, Castillo VC, Papakis V, Leng K, Boggess S, Nowakowski TJ, Wells J, Kampmann M. CRISPRi-based screen of Autism Spectrum Disorder risk genes in microglia uncovers roles of ADNP in microglia endocytosis and synaptic pruning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.01.596962. [PMID: 39605704 PMCID: PMC11601228 DOI: 10.1101/2024.06.01.596962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Autism Spectrum Disorders (ASD) are a set of neurodevelopmental disorders with complex biology. The identification of ASD risk genes from exome-wide association studies and de novo variation analyses has enabled mechanistic investigations into how ASD-risk genes alter development. Most functional genomics studies have focused on the role of these genes in neurons and neural progenitor cells. However, roles for ASD risk genes in other cell types are largely uncharacterized. There is evidence from postmortem tissue that microglia, the resident immune cells of the brain, appear activated in ASD. Here, we used CRISPRi-based functional genomics to systematically assess the impact of ASD risk gene knockdown on microglia activation and phagocytosis. We developed an iPSC-derived microglia-neuron coculture system and high-throughput flow cytometry readout for synaptic pruning to enable parallel CRISPRi-based screening of phagocytosis of beads, synaptosomes, and synaptic pruning. Our screen identified ADNP, a high-confidence ASD risk genes, as a modifier of microglial synaptic pruning. We found that microglia with ADNP loss have altered endocytic trafficking, remodeled proteomes, and increased motility in coculture.
Collapse
Affiliation(s)
- Olivia M Teter
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, CA, USA
| | - Amanda McQuade
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Venus Hagan
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Weiwei Liang
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Nina M Dräger
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Sydney M Sattler
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Brandon B Holmes
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Vincent Cele Castillo
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Vasileios Papakis
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Kun Leng
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA
| | - Steven Boggess
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA
| | - James Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|