1
|
Fang YL, Yen TL, Liu HC, Wang TF, Hsueh YP. Sex-biased zinc responses modulate ribosomal protein expression, protein synthesis and social defects in Cttnbp2 mutant mice. Neurobiol Dis 2025; 211:106932. [PMID: 40300729 DOI: 10.1016/j.nbd.2025.106932] [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: 02/19/2025] [Revised: 04/21/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025] Open
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental conditions influenced by genetic mutations, dietary factors, and sex-specific mechanisms, yet the interplay of these factors remains elusive. Here, we investigate the sex-biased responses of mutant mice carrying an ASD-associated mutation in Cttnbp2 to dietary zinc supplementation using behavioral assays, proteomic and bioinformatic analyses, and puromycin pulse labeling to assess protein synthesis. Our results demonstrate that zinc supplementation enhances ribosomal protein expression and protein synthesis and increases the density and size of dendritic spines in male Cttnbp2 mutant mice, alleviating male-biased social deficits. Analyses of neuronal cultures further revealed that neurons, not astrocytes, respond to zinc to enhance protein synthesis. In contrast, female Cttnbp2 mutants exhibit resilience to differential zinc intake, even under zinc deprivation. Elevated mTOR phosphorylation and increased protein levels of translational initiation factors in female brains may provide a protective mechanism, reducing their sensitivity to zinc deficiency. Cttnbp2 mutations heighten male vulnerability to zinc deprivation, impairing social behaviors. These findings highlight zinc-regulated ribosomal protein expression and protein synthesis as critical mediators of sex-specific ASD phenotypes, offering new insights into dietary interventions.
Collapse
Affiliation(s)
- Yu-Lun Fang
- Institute of Molecular Biology, Academia Sinica, 128, Sec. 2, Academia Rd., Taipei 11529, Taiwan, Republic of China.
| | - Tzu-Li Yen
- Institute of Molecular Biology, Academia Sinica, 128, Sec. 2, Academia Rd., Taipei 11529, Taiwan, Republic of China.
| | - Hou-Cheng Liu
- Institute of Molecular Biology, Academia Sinica, 128, Sec. 2, Academia Rd., Taipei 11529, Taiwan, Republic of China.
| | - Ting-Fang Wang
- Institute of Molecular Biology, Academia Sinica, 128, Sec. 2, Academia Rd., Taipei 11529, Taiwan, Republic of China.
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, 128, Sec. 2, Academia Rd., Taipei 11529, Taiwan, Republic of China.
| |
Collapse
|
2
|
Werren EA, Kalsner L, Ewald J, Peracchio M, King C, Vats P, Audano PA, Robinson PN, Adams MD, Kelly MA, Matson AP. Phenotypic Expansion of Knobloch Syndrome Type 2 in an Individual With a De Novo PAK2 Variant. Am J Med Genet A 2025; 197:e64006. [PMID: 39876536 PMCID: PMC12052494 DOI: 10.1002/ajmg.a.64006] [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: 10/31/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 01/30/2025]
Abstract
P21-activated kinase 2 (PAK2) is a serine/threonine kinase essential for a variety of cellular processes including signal transduction, cellular survival, proliferation, and migration. A recent report proposed monoallelic PAK2 variants cause Knobloch syndrome type 2 (KNO2)-a developmental disorder primarily characterized by ocular anomalies. Here, we identified a novel de novo heterozygous missense variant in PAK2, NM_002577.4:c.1273G>A, p.(D425N), by genome sequencing in an individual with features consistent with KNO2. Notable clinical phenotypes observed in this individual were global developmental delay, congenital retinal detachment, mild cerebral ventriculomegaly, hypotonia, failure to thrive, pyloric stenosis, feeding intolerance, patent ductus arteriosus, and mild facial dysmorphism. The p.(D425N) variant lies within the protein kinase domain and is predicted to be functionally damaging by in silico analysis. Previous clinical genetic testing did not report this variant due to unknown relevance of PAK2 variants at the time of testing, highlighting the importance of reanalysis. Our findings substantiate the candidacy of PAK2 variants in KNO2 and expand the KNO2 clinical phenotypic spectrum.
Collapse
Affiliation(s)
- Elizabeth A. Werren
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, 06032, USA
| | - Louisa Kalsner
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA
- Division of Genetics, Connecticut Children’s Medical Center, Hartford, CT 06106, USA
- Division of Neurology, Connecticut Children’s Medical Center, Hartford, CT 06106, USA
| | - Jessica Ewald
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, 06032, USA
| | - Michael Peracchio
- Division of Genetics, Connecticut Children’s Medical Center, Hartford, CT 06106, USA
| | - Cameron King
- Department of Research, Connecticut Children’s Medical Center, Hartford, CT 06106, USA
| | - Purva Vats
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, 06032, USA
| | - Peter A. Audano
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, 06032, USA
| | - Peter N. Robinson
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, 06032, USA
| | - Mark D. Adams
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, 06032, USA
| | - Melissa A. Kelly
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, 06032, USA
| | - Adam P. Matson
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA
- Division of Neonatology, Connecticut Children’s Medical Center, Hartford, CT 06106, USA
- Department of Immunology, UConn Health, Farmington, Connecticut, 06030, USA
| |
Collapse
|
3
|
Gonzales S, Zhao JZ, Choi NY, Acharya P, Jeong S, Wang X, Lee MY. SOX7: Autism associated gene identified by analysis of multi-Omics data. PLoS One 2025; 20:e0320096. [PMID: 40373085 PMCID: PMC12080844 DOI: 10.1371/journal.pone.0320096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/12/2025] [Indexed: 05/17/2025] Open
Abstract
Genome-wide association studies and next generation sequencing data analyses based on DNA information have identified thousands of mutations associated with autism spectrum disorder (ASD). However, more than 99% of identified mutations are non-coding. Thus, it is unclear which of these mutations might be functional and thus potentially causal variants. Transcriptomic profiling using total RNA-sequencing has been one of the most utilized approaches to link protein levels to genetic information at the molecular level. The transcriptome captures molecular genomic complexity that the DNA sequence solely does not. Some mutations alter a gene's DNA sequence but do not necessarily change expression and/or protein function. To date, few common variants reliably associated with the diagnosis status of ASD despite consistently high estimates of heritability. In addition, reliable biomarkers used to diagnose ASD or molecular mechanisms to define the severity of ASD do not exist. Therefore, it is necessary to integrate DNA and RNA testing together to identify true causal genes and propose useful biomarkers for ASD. We performed gene-based association studies with adaptive test using genome-wide association studies' (GWAS) summary statistics with two large GWAS datasets (ASD 2019 data: 18,382 ASD cases and 27,969 controls [discovery data]; ASD 2017 data: 6,197 ASD cases and 7,377 controls [replication data]) which were obtained from the Psychiatric Genomics Consortium (PGC). In addition, we investigated differential expression between ASD cases and controls for genes identified in gene-based GWAS with two RNA-seq datasets (GSE211154: 20 cases and 19 controls; GSE30573: 3 cases and 3 controls). We identified 5 genes significantly associated with ASD in ASD 2019 data (KIZ-AS1, p = 8.67 × 10-10; KIZ, p = 1.16 × 10-9; XRN2, p = 7.73 × 10-9; SOX7, p = 2.22 × 10-7; LOC101929229 also known as PINX1-DT, p = 2.14 × 10-6). Among these 5 genes, gene SOX7 (p = 0.00087) and LOC101929229 (p = 0.009) were replicated in ASD 2017 data. KIZ-AS1 (p = 0.059) and KIZ (p = 0.06) were close to the boundary of replication in ASD 2017 data. Genes SOX7 (p = 0.036 in all samples; p = 0.044 in white samples) indicated significant expression differences between cases and controls in the GSE211154 RNA-seq data. Furthermore, gene SOX7 was upregulated in cases than in controls in the GSE30573 RNA-seq data (p = 0.0017; Benjamini-Hochberg adjusted p = 0.0085). SOX7 encodes a member of the SOX (SRY-related HMG-box) family of transcription factors pivotally contributing to determining of the cell fate and identity in many lineages. The encoded protein may act as a transcriptional regulator after forming a protein complex with other proteins leading to autism. Gene SOX7 in the transcription factor family could be associated with ASD. This finding may provide new diagnostic and therapeutic strategies for ASD.
Collapse
Affiliation(s)
- Samantha Gonzales
- Department of Biostatistics, Florida International University, Miami, Florida, United States of America
| | - Jane Zizhen Zhao
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Na Young Choi
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, United States of America
| | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, United States of America
| | - Sehoon Jeong
- Department of Artificial Intelligence and Data Science, Sejong University, Seoul, South Korea
| | - Xuexia Wang
- Department of Biostatistics, Florida International University, Miami, Florida, United States of America
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, United States of America
| |
Collapse
|
4
|
Owen MJ, Bray NJ, Walters JTR, O'Donovan MC. Genomics of schizophrenia, bipolar disorder and major depressive disorder. Nat Rev Genet 2025:10.1038/s41576-025-00843-0. [PMID: 40355602 DOI: 10.1038/s41576-025-00843-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
Schizophrenia, bipolar disorder and major depressive disorder - which are the most common adult disorders requiring psychiatric care - contribute substantially to premature mortality and morbidity globally. Treatments for these disorders are suboptimal, there are no diagnostic pathologies or biomarkers and their pathophysiologies are poorly understood. Novel therapeutic and diagnostic approaches are thus badly needed. Given the high heritability of psychiatric disorders, psychiatry has potentially much to gain from the application of genomics to identify molecular risk mechanisms and to improve diagnosis. Recent large-scale, genome-wide association studies and sequencing studies, together with advances in functional genomics, have begun to illuminate the genetic architectures of schizophrenia, bipolar disorder and major depressive disorder and to identify potential biological mechanisms. Genomic findings also point to the aetiological relationships between different diagnoses and to the relationships between adult psychiatric disorders and childhood neurodevelopmental conditions.
Collapse
Affiliation(s)
- Michael J Owen
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.
| | - Nicholas J Bray
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - James T R Walters
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Michael C O'Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
5
|
Tafolla M, Singer H, Lord C. Autism Spectrum Disorder Across the Lifespan. Annu Rev Clin Psychol 2025; 21:193-220. [PMID: 39836874 DOI: 10.1146/annurev-clinpsy-081423-031110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Autism is a neurodevelopmental condition that affects individuals worldwide throughout their lives. Copious advances in research have enhanced our understanding of autism significantly since Dr. Leo Kanner's first description of the condition in 1943. This review aims to provide an overview of our current knowledge of autism, examining its manifestations across age, race, gender, and co-occurring conditions (e.g., intellectual disability) from childhood through adulthood. We also focus on the identification and diagnosis of autism, long-term outcomes with a spotlight on adulthood, and appropriate supports and interventions across different developmental stages for autistic individuals and their families. We stress the importance of a lifespan perspective that considers the evolving needs of individuals with autism as they age, and we highlight the role of longitudinal research.
Collapse
Affiliation(s)
- Maira Tafolla
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California, USA; , ,
| | - Hannah Singer
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California, USA; , ,
| | - Catherine Lord
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California, USA; , ,
| |
Collapse
|
6
|
Wang BM, Mills Z, Jones HF, Montgomery JM, Lee KY. Presymptomatic Biological, Structural, and Functional Diagnostic Biomarkers of Autism Spectrum Disorder. J Neurochem 2025; 169:e70088. [PMID: 40390287 PMCID: PMC12089747 DOI: 10.1111/jnc.70088] [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: 11/29/2024] [Revised: 04/26/2025] [Accepted: 05/06/2025] [Indexed: 05/21/2025]
Abstract
Autism spectrum disorder (ASD) is a common neurodevelopmental disorder clinically diagnosed by persistent deficits in three areas of social communication and interaction, plus at least two of four types of restricted repetitive behaviors. ASD has been shown to be caused by genetic predisposition and environmental factors; however, the heterogeneity of ASD complicates its diagnosis and treatment. Early behavioral interventions have shown significant benefits, emphasizing the urgent need for reliable diagnostic biomarkers to enhance long-term outcomes. Here we provide a systematic review that outlines current findings on genetic and neurological biomarkers for presymptomatic ASD diagnoses, assessed prior to the observation of behavioral manifestations. Specifically, we offer insights into the mechanisms of presymptomatic neurological, biological, structural, and functional markers for ASD, compare outcomes across studies, and critically assess their limitations and implications. Recent findings highlight genotype-guided therapeutic strategies in animal models, such as dietary zinc supplementation for reversing ASD-associated behaviors by synaptic deficits. However, the differential efficacy based on underlying genotypes, along with challenges in identifying reliable genomic biomarkers prior to symptom onset, indicates the need for further research. Notably, recent advancements in imaging technologies like magnetic resonance imaging, electroencephalography, and pupillometry have shown promising markers in neonates, and at 3 and 9 months old, respectively. Newer developments in magnetoencephalography hardware can facilitate the much-needed infant ASD studies. It is important to note that many of these biomarker findings are preliminary, and further validation for clinical use is required. Continued research is needed to advance the practicality, reliability, and acceptability of these biomarkers to improve ASD diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Bonnie M. Wang
- Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Zoe Mills
- Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
- Centre for Brain Research, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Hannah F. Jones
- Centre for Brain Research, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
- Department of NeuroservicesStarship Children's HospitalAucklandNew Zealand
| | - Johanna M. Montgomery
- Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
- Centre for Brain Research, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Kevin Y. Lee
- Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
- Centre for Brain Research, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
7
|
Budisteanu M, Papuc SM, Erbescu A, Glangher A, Andrei E, Rad F, Hinescu ME, Arghir A. Review of structural neuroimaging and genetic findings in autism spectrum disorder - a clinical perspective. Rev Neurosci 2025; 36:295-314. [PMID: 39566028 DOI: 10.1515/revneuro-2024-0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/03/2024] [Indexed: 11/22/2024]
Abstract
Autism spectrum disorders (ASDs) are neurodevelopmental conditions characterized by deficits in social relationships and communication and restrictive, repetitive behaviors and interests. ASDs form a heterogeneous group from a clinical and genetic perspective. Currently, ASDs diagnosis is based on the clinical observation of the individual's behavior. The subjective nature of behavioral diagnoses, in the context of ASDs heterogeneity, contributes to significant variation in the age at ASD diagnosis. Early detection has been proved to be critical in ASDs, as early start of appropriate therapeutic interventions greatly improve the outcome for some children. Structural magnetic resonance imaging (MRI) is widely used in the diagnostic work-up of neurodevelopmental conditions, including ASDs, mostly for brain malformations detection. Recently, the focus of brain imaging shifted towards quantitative MRI parameters, aiming to identify subtle changes that may establish early detection biomarkers. ASDs have a strong genetic component; deletions and duplications of several genomic loci have been strongly associated with ASDs risk. Consequently, a multitude of neuroimaging and genetic findings emerged in ASDs in the recent years. The association of gross or subtle changes in brain morphometry and volumes with different genetic defects has the potential to bring new insights regarding normal development and pathomechanisms of various disorders affecting the brain. Still, the clinical implications of these discoveries and the impact of genetic abnormalities on brain structure and function are unclear. Here we review the literature on brain imaging correlated with the most prevalent genomic imbalances in ASD, and discuss the potential clinical impact.
Collapse
Affiliation(s)
- Magdalena Budisteanu
- Alexandru Obregia Clinical Hospital of Psychiatry, 041914, Soseaua Berceni 10, Bucharest, Romania
- Victor Babes National Institute of Pathology, 050096, Splaiul Independentei 99-101, Bucharest, Romania
- Faculty of Medicine, Titu Maiorescu University, 031593, Calea Vacaresti 187, Bucharest, Romania
| | - Sorina Mihaela Papuc
- Victor Babes National Institute of Pathology, 050096, Splaiul Independentei 99-101, Bucharest, Romania
| | - Alina Erbescu
- Victor Babes National Institute of Pathology, 050096, Splaiul Independentei 99-101, Bucharest, Romania
| | - Adelina Glangher
- Alexandru Obregia Clinical Hospital of Psychiatry, 041914, Soseaua Berceni 10, Bucharest, Romania
| | - Emanuela Andrei
- Alexandru Obregia Clinical Hospital of Psychiatry, 041914, Soseaua Berceni 10, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, 050474, Bulevardul Eroii Sanitari 8, Bucharest, Romania
| | - Florina Rad
- Alexandru Obregia Clinical Hospital of Psychiatry, 041914, Soseaua Berceni 10, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, 050474, Bulevardul Eroii Sanitari 8, Bucharest, Romania
| | - Mihail Eugen Hinescu
- Victor Babes National Institute of Pathology, 050096, Splaiul Independentei 99-101, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, 050474, Bulevardul Eroii Sanitari 8, Bucharest, Romania
| | - Aurora Arghir
- Victor Babes National Institute of Pathology, 050096, Splaiul Independentei 99-101, Bucharest, Romania
| |
Collapse
|
8
|
La Monica I, Di Iorio MR, Sica A, Rufino F, Sotira C, Pastore L, Lombardo B. Autism Spectrum Disorder: Genetic Mechanisms and Inheritance Patterns. Genes (Basel) 2025; 16:478. [PMID: 40428300 PMCID: PMC12111747 DOI: 10.3390/genes16050478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/18/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that first develops in early childhood and is characterized by restricted interests, activities, and behaviors, as well as difficulties with social interactions and communication. ASD arises from a complex interaction between environmental factors and genetic inheritance, influenced by epigenetic mechanisms. With an estimated heritability of 70-90%, ASD is highly familial, indicating that genetic factors play a significant role in its development. This shows how hundreds of genetic variants contribute to ASD, whose risk effects are highly variable and are often related to other conditions; these genetic alterations are at different levels, which include single gene mutations, monogenic disorders, genomic variants, and chromosomal abnormalities. Copy number variants (CNVs) appear to contribute significantly to understanding the pathogenesis of this complex disease. In some cases, single CNVs in genomic DNA are pathogenic and causative, supporting the hypothesis that some sporadic cases of ASD may result from rare mutations with significant clinical impact. However, in many cases, there are common genomic variants that increase the risk of developing ASD but are insufficient by themselves to determine an ASD phenotype, and rare genomic variants, of various sizes, inherited from a parent or de novo, that can be associated with the ASD phenotype. Therefore, the aim of this review is to deepen the concept of ASD inheritance through the two-hit theory of CNVs, in which the concomitant presence of two alterations could determine the clinical phenotypes, the concept of incomplete penetrance for inherited CNVs with pathogenic clinical significance, and the presence of compound heterozygosity. These aspects represent important mechanisms underlying the pathogenesis of autism, contributing to a better elucidation for the understanding of the genetic contribution to the ASD phenotype.
Collapse
Affiliation(s)
- Ilaria La Monica
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy; (I.L.M.); (A.S.); (F.R.); (C.S.); (L.P.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy;
| | - Maria Rosaria Di Iorio
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy;
| | - Antonia Sica
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy; (I.L.M.); (A.S.); (F.R.); (C.S.); (L.P.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy;
| | - Francesca Rufino
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy; (I.L.M.); (A.S.); (F.R.); (C.S.); (L.P.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy;
| | - Chiara Sotira
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy; (I.L.M.); (A.S.); (F.R.); (C.S.); (L.P.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy;
| | - Lucio Pastore
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy; (I.L.M.); (A.S.); (F.R.); (C.S.); (L.P.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy;
| | - Barbara Lombardo
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy; (I.L.M.); (A.S.); (F.R.); (C.S.); (L.P.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy;
| |
Collapse
|
9
|
Mei L, Hu C, Jin G, Ge C, Zhu Y, Li D, Peng W, Li H, Xu X, Jiang Y, Xu G, Xu Q. Molecular genetic testing and cohort analysis of 32 twin pairs with neurodevelopmental disorders-Reporting a novel de novo variant of TET3. Hum Genomics 2025; 19:42. [PMID: 40259394 PMCID: PMC12013076 DOI: 10.1186/s40246-025-00748-3] [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: 10/20/2024] [Accepted: 03/30/2025] [Indexed: 04/23/2025] Open
Abstract
Neurodevelopmental disorders (NDDs) pose significant challenges due to their impact on cognitive, social and motor abilities, often rooted in genetic factors such as copy number variations (CNVs) and single nucleotide variantions (SNVs). Molecular genetic testing, advanced due to sequencing technologies, is instrumental in diagnosing NDDs, with twins offering unique perspectives in detecting novel de novo CNVs and SNVs. The study enrolled 32 pairs of twins that underwent molecular genetic testing and comprehensive clinical data collection. Additionally, we analyzed the potential deleterious effects of a novel de novo TET methylcytosine dioxygenase 3 (TET3) variant (c.4927G > A) using western blotting, immunofluorescence assay and enzymatic activity assay. Analyzing simultaneously, the overall detection yield of molecular genetic testing was 17.2% (11/64). Children with disease-related genetic variants had lower total developmental quotients (DQ) than children without disease-related genetic variants. One pair of monozygotic twins carried a novel de novo TET3 variant. Immunostaining assay revealed that while the wildtype TET3 protein was evenly distributed in the nucleus, the variant was concentrated around the nucleus. Anenzymatic assay using corresponding TET2 mutants suggested that the variant has a significantly reduced activity. Taken together, our study elaborated molecular genetic testing results of 32 pairs of twins and found that children with lower developmental levels are prone to possessing identifiable genetic variants. We reported the clinical phenotype of a pair of monozygotic twins carrying a novel de novo TET3 variant and confirmed the detrimental effects of this variant in vitro.
Collapse
Affiliation(s)
- Lianni Mei
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Chunchun Hu
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Guangbo Jin
- Institutes of Biomedical Sciences, Shanghai Xuhui Central Hospital, Medical College of Fudan University, Chinese Academy of Medical Sciences (RU069), Shanghai, China
| | - Chuanhui Ge
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontier Center for Brain Science, Fudan University, Shanghai, China
| | - Yiting Zhu
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Dongyun Li
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Wenzhu Peng
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontier Center for Brain Science, Fudan University, Shanghai, China
| | - Huiping Li
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Xiu Xu
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Yan Jiang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontier Center for Brain Science, Fudan University, Shanghai, China
| | - Guoliang Xu
- Institutes of Biomedical Sciences, Shanghai Xuhui Central Hospital, Medical College of Fudan University, Chinese Academy of Medical Sciences (RU069), Shanghai, China.
- China State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.
| | - Qiong Xu
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China.
- Department of Child Health Care, Anhui Provincial Children's Hospital, Hefei, China.
| |
Collapse
|
10
|
Kumar K, Liao Z, Kopal J, Moreau C, Ching CRK, Modenato C, Snyder W, Kazem S, Martin CO, Bélanger AM, Fontaine VK, Jizi K, Boen R, Huguet G, Saci Z, Kushan L, Silva AI, van den Bree MBM, Linden DEJ, Owen MJ, Hall J, Lippé S, Dumas G, Draganski B, Almasy L, Thomopoulos SI, Jahanshad N, Sønderby IE, Andreassen OA, Glahn DC, Raznahan A, Bearden CE, Paus T, Thompson PM, Jacquemont S. Cortical differences across psychiatric disorders and associated common and rare genetic variants. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.16.25325971. [PMID: 40321288 PMCID: PMC12047953 DOI: 10.1101/2025.04.16.25325971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Genetic studies have identified common and rare variants increasing the risk for neurodevelopmental and psychiatric disorders (NPDs). These risk variants have also been shown to influence the structure of the cerebral cortex. However, it is unknown whether cortical differences associated with genetic variants are linked to the risk they confer for NPDs. To answer this question, we analyzed cortical thickness (CT) and surface area (SA) for common and rare variants associated with NPDs, in ~33000 individuals from the general population and clinical cohorts, as well as ENIGMA summary statistics for 8 NPDs. Rare and common genetic variants increasing risk for NPDs were preferentially associated with total SA, while NPDs were preferentially associated with mean CT. Larger effects on mean CT, but not total SA, were observed in NPD medicated subgroups. At the regional level, genetic variants were preferentially associated with effects in sensorimotor areas, while NPDs showed higher effects in association areas. We show that schizophrenia- and bipolar-disorder-associated SNPs show positive and negative effect sizes on SA suggesting that their aggregated effects cancel out in additive polygenic models. Overall, CT and SA differences associated with NPDs do not relate to those observed across individual genetic variants and may be linked with critical non-genetic factors, such as medication and the lived experience of the disorder.
Collapse
Affiliation(s)
- Kuldeep Kumar
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
| | - Zhijie Liao
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
| | - Jakub Kopal
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Clara Moreau
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
| | - Christopher R K Ching
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Claudia Modenato
- LREN - Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Switzerland
| | - Will Snyder
- Section on Developmental Neurogenomics, Human Genetics Branch, NIMH, NIH, Bethesda, MD, USA
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Sayeh Kazem
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
| | | | | | - Valérie K Fontaine
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
| | - Khadije Jizi
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
| | - Rune Boen
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Psychology, UCLA, Los Angeles, USA
| | - Guillaume Huguet
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
| | - Zohra Saci
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
| | - Leila Kushan
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Psychology, UCLA, Los Angeles, USA
| | - Ana I Silva
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, MN, USA
| | - Marianne B M van den Bree
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - David E J Linden
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- Mental Health and Neuroscience Research Institute, Maastricht University, Netherlands
| | - Michael J Owen
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Jeremy Hall
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Sarah Lippé
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
| | - Guillaume Dumas
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
| | - Bogdan Draganski
- LREN - Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Switzerland
- Neurology Department, Max-Planck-Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Department of Neurology, Inselspital, University of Bern, Bern, Switzerland
- University Institute for Diagnostic and Interventional Neuroradiology, Inselspital, University of Bern, Bern, Switzerland
| | - Laura Almasy
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, PA, USA
- Lifespan Brain Institute, Children's Hospital of Philadelphia and Penn Medicine, PA, USA
- Department of Genetics, University of Pennsylvania, PA, USA
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Ida E Sønderby
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - David C Glahn
- Harvard Medical School, Department of Psychiatry, 25 Shattuck St, Boston, MA, USA
- Boston Children's Hospital, Tommy Fuss Center for Neuropsychiatric Disease Research, 300 Longwood Avenue, Boston, MA, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, NIMH, NIH, Bethesda, MD, USA
| | - Carrie E Bearden
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Psychology, UCLA, Los Angeles, USA
| | - Tomas Paus
- Centre de recherche CHU Sainte-Justine and University of Montreal, Canada
- Departments of Psychiatry and Neuroscience, University of Montreal, Montreal, Quebec, Canada
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | | |
Collapse
|
11
|
Mei Y, Gosztyla ML, Tan X, Dozier LE, Wilkinson B, McKetney J, Lee J, Chen M, Tsai D, Kopalle H, Gritsenko MA, Hartel N, Graham NA, Flores I, Gilmore-Hall SK, Xu S, Marquez CA, Liu SN, Fong D, Chen J, Licon K, Hong D, Wright SN, Kreisberg JF, Nott A, Smith RD, Qian WJ, Swaney DL, Iakoucheva LM, Krogan NJ, Patrick GN, Zhou Y, Feng G, Coba MP, Yeo GW, Ideker T. Integrated multi-omic characterizations of the synapse reveal RNA processing factors and ubiquitin ligases associated with neurodevelopmental disorders. Cell Syst 2025; 16:101204. [PMID: 40054464 DOI: 10.1016/j.cels.2025.101204] [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: 05/03/2024] [Revised: 11/26/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025]
Abstract
The molecular composition of the excitatory synapse is incompletely defined due to its dynamic nature across developmental stages and neuronal populations. To address this gap, we apply proteomic mass spectrometry to characterize the synapse in multiple biological models, including the fetal human brain and human induced pluripotent stem cell (hiPSC)-derived neurons. To prioritize the identified proteins, we develop an orthogonal multi-omic screen of genomic, transcriptomic, interactomic, and structural data. This data-driven framework identifies proteins with key molecular features intrinsic to the synapse, including characteristic patterns of biophysical interactions and cross-tissue expression. The multi-omic analysis captures synaptic proteins across developmental stages and experimental systems, including 493 synaptic candidates supported by proteomics. We further investigate three such proteins that are associated with neurodevelopmental disorders-Cullin 3 (CUL3), DEAD-box helicase 3 X-linked (DDX3X), and Y-box binding protein-1 (YBX1)-by mapping their networks of physically interacting synapse proteins or transcripts. Our study demonstrates the potential of an integrated multi-omic approach to more comprehensively resolve the synaptic architecture.
Collapse
Affiliation(s)
- Yuan Mei
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92023, USA
| | - Maya L Gosztyla
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Sanford Stem Cell Institute Innovation Center, University of California, San Diego, La Jolla, CA 92037, USA; Center for RNA Technologies and Therapeutics, University of California, San Diego, La Jolla, CA, USA
| | - Xinzhu Tan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 1A1, Canada
| | - Lara E Dozier
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brent Wilkinson
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - Justin McKetney
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA; University of California, San Francisco, Quantitative Biosciences Institute, San Francisco, CA 94158, USA; University of California, San Francisco, Department of Cellular and Molecular Pharmacology, San Francisco, CA 94143, USA
| | - John Lee
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael Chen
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dorothy Tsai
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hema Kopalle
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92023, USA
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Nicolas Hartel
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Nicholas A Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Ilse Flores
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Stephen K Gilmore-Hall
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shuhao Xu
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Sanford Stem Cell Institute Innovation Center, University of California, San Diego, La Jolla, CA 92037, USA; Center for RNA Technologies and Therapeutics, University of California, San Diego, La Jolla, CA, USA
| | - Charlotte A Marquez
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sophie N Liu
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dylan Fong
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jing Chen
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kate Licon
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Derek Hong
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sarah N Wright
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jason F Kreisberg
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Stem Cell Institute Innovation Center, University of California, San Diego, La Jolla, CA 92037, USA
| | - Alexi Nott
- Department of Brain Sciences, Imperial College London, White City Campus, London W12 7RH, UK; UK Dementia Research Institute, Imperial College London, White City Campus, London W12 0BZ, UK
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Danielle L Swaney
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA; University of California, San Francisco, Quantitative Biosciences Institute, San Francisco, CA 94158, USA; University of California, San Francisco, Department of Cellular and Molecular Pharmacology, San Francisco, CA 94143, USA
| | - Lilia M Iakoucheva
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nevan J Krogan
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA; University of California, San Francisco, Quantitative Biosciences Institute, San Francisco, CA 94158, USA; University of California, San Francisco, Department of Cellular and Molecular Pharmacology, San Francisco, CA 94143, USA
| | - Gentry N Patrick
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yang Zhou
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 1A1, Canada
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Marcelo P Coba
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA.
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92023, USA; Sanford Stem Cell Institute Innovation Center, University of California, San Diego, La Jolla, CA 92037, USA; Center for RNA Technologies and Therapeutics, University of California, San Diego, La Jolla, CA, USA.
| | - Trey Ideker
- Division of Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
12
|
Dubois AEE, Audet-Duchesne E, Knoth IS, Martin CO, Jizi K, Tamer P, Younis N, Jacquemont S, Dumas G, Lippé S. Genetic modulation of brain dynamics in neurodevelopmental disorders: the impact of copy number variations on resting-state EEG. Transl Psychiatry 2025; 15:139. [PMID: 40216767 PMCID: PMC11992136 DOI: 10.1038/s41398-025-03324-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 01/21/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
Research has shown that many copy number variations (CNVs) increase the risk of neurodevelopmental disorders (e.g., autism, ADHD, schizophrenia). However, little is known about the effects of CNVs on brain development and function. Resting-state electroencephalography (EEG) is a suitable method to study the disturbances of neuronal functioning in CNVs. We aimed to determine whether there are resting-state EEG signatures that are characteristic of children with pathogenic CNVs. EEG resting-state brain activity of 109 CNV carriers (66 deletion carriers, 43 duplication carriers) aged 3 to 17 years was recorded for 4 minutes. To better account for developmental variations, EEG indices (power spectral density and functional connectivity) were corrected with a normative model estimated from 256 Healthy Brain Network controls. Results showed a decreased exponent of the aperiodic activity and a reduced alpha peak frequency in CNV carriers. Additionally, the study showed altered periodic components and connectivity in several frequency bands. Deletion and duplication carriers exhibited a similar overall pattern of deviations in spectral and connectivity measures, although the significance and effect sizes relative to the control group varied across frequency bands. Deletion and duplication carriers can be differentiated by their periodic power in the gamma band and connectivity in the low alpha band, with duplication carriers showing more disrupted alterations than deletion carriers. The distinctive alterations in spectral patterns were found to be most prominent during adolescence. The results suggest that CNV carriers show electrophysiological alterations compared to neurotypical controls, regardless of the gene dosage effect and their affected genomic region. At the same time, while duplications and deletions share common electrophysiological alterations, each exhibits distinct brain alteration signatures that reflect gene dosage-specific effects.
Collapse
Affiliation(s)
- Adrien E E Dubois
- Research Center of the Sainte-Justine Mother and Child University Hospital Center (CHU Sainte-Justine), Montreal, QC, H3T 1C5, Canada
- Department of Neurosciences, University of Montreal, Montreal, QC, H3C 3J7, Canada
| | - Elisabeth Audet-Duchesne
- Research Center of the Sainte-Justine Mother and Child University Hospital Center (CHU Sainte-Justine), Montreal, QC, H3T 1C5, Canada
- Department of Psychology, University of Montreal, Montreal, QC, H2V 2S9, Canada
| | - Inga Sophia Knoth
- Research Center of the Sainte-Justine Mother and Child University Hospital Center (CHU Sainte-Justine), Montreal, QC, H3T 1C5, Canada
| | - Charles-Olivier Martin
- Research Center of the Sainte-Justine Mother and Child University Hospital Center (CHU Sainte-Justine), Montreal, QC, H3T 1C5, Canada
| | - Khadije Jizi
- Research Center of the Sainte-Justine Mother and Child University Hospital Center (CHU Sainte-Justine), Montreal, QC, H3T 1C5, Canada
| | - Petra Tamer
- Research Center of the Sainte-Justine Mother and Child University Hospital Center (CHU Sainte-Justine), Montreal, QC, H3T 1C5, Canada
| | - Nadine Younis
- Research Center of the Sainte-Justine Mother and Child University Hospital Center (CHU Sainte-Justine), Montreal, QC, H3T 1C5, Canada
| | - Sébastien Jacquemont
- Research Center of the Sainte-Justine Mother and Child University Hospital Center (CHU Sainte-Justine), Montreal, QC, H3T 1C5, Canada
- Department of Pediatrics, University de Montreal, Montreal, QC, H3T 1C5, Canada
| | - Guillaume Dumas
- Research Center of the Sainte-Justine Mother and Child University Hospital Center (CHU Sainte-Justine), Montreal, QC, H3T 1C5, Canada.
- Department of Psychiatry and Addictology, University of Montreal, Montreal, QC, H3T 1J4, Canada.
- Mila - Québec AI Institute, University of Montreal, Montreal, QC, Canada.
| | - Sarah Lippé
- Research Center of the Sainte-Justine Mother and Child University Hospital Center (CHU Sainte-Justine), Montreal, QC, H3T 1C5, Canada.
- Department of Psychology, University of Montreal, Montreal, QC, H2V 2S9, Canada.
| |
Collapse
|
13
|
He T, Zhang M, Qin J, Wang Y, Li S, Du C, Jiao J, Ji F. Endothelial PD-1 Regulates Vascular Homeostasis and Oligodendrogenesis during Brain Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417410. [PMID: 40013943 PMCID: PMC12021089 DOI: 10.1002/advs.202417410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Appropriate vascular and neural development is essential for central nervous system (CNS). Although programmed cell death receptor 1 (PD-1) mediates neurogenesis, its role in cerebrovascular development remains poorly understood. Here, a correlation between cerebral vessels and oligodendrocyte precursor cells (OPCs) is revealed during brain development. The ablation of endothelial PD-1 triggers cortical hypervascularization through excessive angiogenic sprouting, concomitantly driving OPC differentiation. These alterations disrupt blood brain barrier (BBB) maturation, induce dysmyelination, and ultimately result in abnormal behavior in mice. Mechanistically, the loss of endothelial PD-1 suppresses the activity of the Wnt/β-catenin signaling pathway, thereby disrupting normal angiogenesis. Concurrently, it activates the MEK1/2-ERK1/2-GLI1 pathway, leading to increased GREMLIN1 (GREM1) expression. Elevated GREM1 secretion inhibits the BMP/SMAD1/5/SMAD4 signaling cascade in OPCs, which inhibits oligodendrogenesis and myelination. These findings indicate the importance of endothelial cell-intrinsic PD-1 in regulating the oligovascular niche, and suggest potential therapeutic implications for neurological disorders associated with disrupted vascular development.
Collapse
Affiliation(s)
- Tingting He
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Sino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing100190China
| | - Mengtian Zhang
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jie Qin
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yanyan Wang
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Sihan Li
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Chaoyi Du
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jianwei Jiao
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Fen Ji
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| |
Collapse
|
14
|
Yang J, Ma G, Du X, Xie J, Wang M, Wang W, Guo B, Wu S. Deciphering the Role of Shank3 in Dendritic Morphology and Synaptic Function Across Postnatal Developmental Stages in the Shank3B KO Mouse. Neurosci Bull 2025; 41:583-599. [PMID: 39693031 PMCID: PMC11978597 DOI: 10.1007/s12264-024-01330-y] [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: 05/10/2024] [Accepted: 09/14/2024] [Indexed: 12/19/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is marked by early-onset neurodevelopmental anomalies, yet the temporal dynamics of genetic contributions to these processes remain insufficiently understood. This study aimed to elucidate the role of the Shank3 gene, known to be associated with monogenic causes of autism, in early developmental processes to inform the timing and mechanisms for potential interventions for ASD. Utilizing the Shank3B knockout (KO) mouse model, we examined Shank3 expression and its impact on neuronal maturation through Golgi staining for dendritic morphology and electrophysiological recordings to measure synaptic function in the anterior cingulate cortex (ACC) across different postnatal stages. Our longitudinal analysis revealed that, while Shank3B KO mice displayed normal neuronal morphology at one week postnatal, significant impairments in dendritic growth and synaptic activity emerged by two to three weeks. These findings highlight the critical developmental window during which Shank3 is essential for neuronal and synaptic maturation in the ACC.
Collapse
Affiliation(s)
- Jing Yang
- Department of Basic Medicine, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Guaiguai Ma
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaohui Du
- Department of Basic Medicine, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Jinyi Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Mengmeng Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Baolin Guo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
- Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
15
|
Gao S, Shan C, Zhang R, Wang T. Genetic advances in neurodevelopmental disorders. MEDICAL REVIEW (2021) 2025; 5:139-151. [PMID: 40224365 PMCID: PMC11987507 DOI: 10.1515/mr-2024-0040] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/14/2024] [Indexed: 04/15/2025]
Abstract
Neurodevelopmental disorders (NDDs) are a group of highly heterogeneous diseases that affect children's social, cognitive, and emotional functioning. The etiology is complicated with genetic factors playing an important role. During the past decade, large-scale whole exome sequencing (WES) and whole genome sequencing (WGS) have vastly advanced the genetic findings of NDDs. Various forms of variants have been reported to contribute to NDDs, such as de novo mutations (DNMs), copy number variations (CNVs), rare inherited variants (RIVs), and common variation. By far, over 200 high-risk NDD genes have been identified, which are involved in biological processes including synaptic function, transcriptional and epigenetic regulation. In addition, monogenic, oligogenic, polygenetic, and omnigenic models have been proposed to explain the genetic architecture of NDDs. However, the majority of NDD patients still do not have a definitive genetic diagnosis. In the future, more types of risk factors, as well as noncoding variants, are await to be identified, and including their interplay mechanisms are key to resolving the etiology and heterogeneity of NDDs.
Collapse
Affiliation(s)
- Shilin Gao
- Department of Neuroscience, Neuroscience Research Institute, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, China
| | - Chaoyi Shan
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Rong Zhang
- Department of Neuroscience, Neuroscience Research Institute, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, China
| | - Tianyun Wang
- Department of Neuroscience, Neuroscience Research Institute, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, China
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Autism Research Center, Peking University Health Science Center, Beijing, China
| |
Collapse
|
16
|
Sun Z, Zhu H, He X, Lendemeijer B, Wang Z, Fan J, Sun Y, Zhang Z, Markx S, Kushner SA, Xu B, Gogos JA. Genomic and Transcriptomic Signatures of SETD1A Disruption in Human Excitatory Neuron Development and Psychiatric Disease Risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645419. [PMID: 40196527 PMCID: PMC11974865 DOI: 10.1101/2025.03.26.645419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Genetic disruption of SETD1A markedly increases the risk for schizophrenia. To elucidate the underlying mechanisms, we generated isogenic organoid models of the developing human cerebral cortex harboring a SETD1A loss-of-function schizophrenia risk mutation. Employing chromatin profiling combined with RNA sequencing, we identified high-confidence SETD1A target genes, analyzed the impact of the mutation on SETD1A binding and transcriptional regulation and validated key findings with orthogonal approaches. Disruption of SETD1A function disturbs the finely tuned temporal gene expression in the excitatory neuron lineage, yielding an aberrant transcriptional program that compromises key regulatory and metabolic pathways essential for neurodevelopmental transitions. Although overall SETD1A binding remains unchanged in mutant neurons, we identified localized alterations in SETD1A binding that correlate with shifts in H3K4me3 levels and gene expression. These changes are enriched at enhancer regions, suggesting that enhancer-regulated genes are especially vulnerable to SETD1A reduction. Notably, target genes with enhancer-bound SETD1A are primarily linked to neuronal functions while those with promoter-bound SETD1A are enriched for basic cellular functions. By mapping the SETD1A binding landscape in excitatory neurons of the human fetal frontal cortex and integrating multimodal neuroimaging and genetic datasets, we demonstrate that the genomic context of SETD1A binding differentially correlates with macroscale brain organization and establish a link between SETD1A-bound enhancers, schizophrenia-associated brain alterations and genetic susceptibility. Our study advances our understanding of the role of SETD1A binding patterns in schizophrenia pathogenesis, offering insights that may guide future therapeutic strategies.
Collapse
Affiliation(s)
- Zhixiong Sun
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Huixiang Zhu
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Xiaofu He
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
| | - Bas Lendemeijer
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Zanxu Wang
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
| | - Jack Fan
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
| | - Yan Sun
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Zhiguo Zhang
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - Sander Markx
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Steven A. Kushner
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Joseph A. Gogos
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Neuroscience, Columbia University, New York, NY 10032, USA
| |
Collapse
|
17
|
Ashitomi H, Nakagawa T, Nakagawa M, Hosoi T. Cullin-RING Ubiquitin Ligases in Neurodevelopment and Neurodevelopmental Disorders. Biomedicines 2025; 13:810. [PMID: 40299365 PMCID: PMC12024872 DOI: 10.3390/biomedicines13040810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Ubiquitination is a dynamic and tightly regulated post-translational modification essential for modulating protein stability, trafficking, and function to preserve cellular homeostasis. This process is orchestrated through a hierarchical enzymatic cascade involving three key enzymes: the E1 ubiquitin-activating enzyme, the E2 ubiquitin-conjugating enzyme, and the E3 ubiquitin ligase. The final step of ubiquitination is catalyzed by the E3 ubiquitin ligase, which facilitates the transfer of ubiquitin from the E2 enzyme to the substrate, thereby dictating which proteins undergo ubiquitination. Emerging evidence underscores the critical roles of ubiquitin ligases in neurodevelopment, regulating fundamental processes such as neuronal polarization, axonal outgrowth, synaptogenesis, and synaptic function. Mutations in genes encoding ubiquitin ligases and the consequent dysregulation of these pathways have been increasingly implicated in a spectrum of neurodevelopmental disorders, including autism spectrum disorder, intellectual disability, and attention-deficit/hyperactivity disorder. This review synthesizes current knowledge on the molecular mechanisms underlying neurodevelopment regulated by Cullin-RING ubiquitin ligases-the largest subclass of ubiquitin ligases-and their involvement in the pathophysiology of neurodevelopmental disorders. A deeper understanding of these mechanisms holds significant promise for informing novel therapeutic strategies, ultimately advancing clinical outcomes for individuals affected by neurodevelopmental disorders.
Collapse
Affiliation(s)
- Honoka Ashitomi
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0084, Japan; (H.A.)
| | - Tadashi Nakagawa
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0084, Japan; (H.A.)
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Makiko Nakagawa
- Institute of Gene Research, Yamaguchi University Science Research Center, Ube 755-8505, Japan
- Advanced Technology Institute, Life Science Division, Yamaguchi University, Ube 755-8611, Japan
| | - Toru Hosoi
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0084, Japan; (H.A.)
| |
Collapse
|
18
|
Weekley BH, Ahmed NI, Maze I. Elucidating neuroepigenetic mechanisms to inform targeted therapeutics for brain disorders. iScience 2025; 28:112092. [PMID: 40160416 PMCID: PMC11951040 DOI: 10.1016/j.isci.2025.112092] [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] [Indexed: 04/02/2025] Open
Abstract
The evolving field of neuroepigenetics provides important insights into the molecular foundations of brain function. Novel sequencing technologies have identified patient-specific mutations and gene expression profiles involved in shaping the epigenetic landscape during neurodevelopment and in disease. Traditional methods to investigate the consequences of chromatin-related mutations provide valuable phenotypic insights but often lack information on the biochemical mechanisms underlying these processes. Recent studies, however, are beginning to elucidate how structural and/or functional aspects of histone, DNA, and RNA post-translational modifications affect transcriptional landscapes and neurological phenotypes. Here, we review the identification of epigenetic regulators from genomic studies of brain disease, as well as mechanistic findings that reveal the intricacies of neuronal chromatin regulation. We then discuss how these mechanistic studies serve as a guideline for future neuroepigenetics investigations. We end by proposing a roadmap to future therapies that exploit these findings by coupling them to recent advances in targeted therapeutics.
Collapse
Affiliation(s)
- Benjamin H. Weekley
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Newaz I. Ahmed
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
19
|
Abdallah SB, Olfson E, Cappi C, Greenspun S, Zai G, Rosário MC, Willsey AJ, Shavitt RG, Miguel EC, Kennedy JL, Richter MA, Fernandez TV. Characterizing Rare DNA Copy-Number Variants in Pediatric Obsessive-Compulsive Disorder. J Am Acad Child Adolesc Psychiatry 2025:S0890-8567(25)00160-1. [PMID: 40122455 DOI: 10.1016/j.jaac.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 01/21/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE Pediatric obsessive-compulsive disorder (OCD) is a common neuropsychiatric disorder in which genetic factors play an important role. Recent studies have demonstrated an enrichment of rare de novo DNA single-nucleotide variants in persons with OCD compared to controls, and larger studies have examined copy-number variants (CNVs) using microarray data. Our study examines rare de novo CNVs using whole-exome sequencing (WES) data to provide additional insight into genetic factors and biological processes underlying OCD. METHOD We detected CNVs using whole-exome DNA sequencing (WES) data from 183 OCD trio families (unaffected parents and children with OCD) and 771 control families to test the hypothesis that rare de novo CNVs are enriched in persons with OCD compared to controls. Our primary analysis used the eXome-Hidden Markov Model (XHMM) to identify CNVs in silico. We performed burden analyses comparing persons with OCD vs controls and downstream biological systems analyses of CNVs in probands with OCD. We then used a second algorithm (GATK-gCNV) to confirm our primary analysis. RESULTS Our findings demonstrate a higher rate of rare de novo CNVs detected by WES in persons with OCD (0.07 CNVs per proband) compared to controls (0.005) (corrected rate ratio = 11.7 95% CI = 3.6-50.0, p = 4.00×10-6). We confirmed this enrichment using GATK-gCNV. The majority of these rare de novo CNVs in persons with OCD are predicted to be pathogenic or likely pathogenic, and an examination of genes disrupted by rare de novo CNVs in persons with OCD finds enrichment of several Gene Ontology sets. CONCLUSION This study shows for the first time an enrichment of rare de novo CNVs detected by WES in OCD, complementing previous, larger CNV studies and providing additional insight into genetic factors underlying OCD risk.
Collapse
Affiliation(s)
| | - Emily Olfson
- Yale University School of Medicine, New Haven, Connecticut
| | - Carolina Cappi
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Gwyneth Zai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - A Jeremy Willsey
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California
| | - Roseli G Shavitt
- Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - James L Kennedy
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Margaret A Richter
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Frederick W. Thompson Anxiety Disorders Centre, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | | |
Collapse
|
20
|
Bertollo AG, Puntel CF, da Silva BV, Martins M, Bagatini MD, Ignácio ZM. Neurobiological Relationships Between Neurodevelopmental Disorders and Mood Disorders. Brain Sci 2025; 15:307. [PMID: 40149827 PMCID: PMC11940368 DOI: 10.3390/brainsci15030307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
According to the Diagnostic and Statistical Manual of Mental Disorders (DSM-5), neurodevelopmental disorders (NDDs) are a group of conditions that arise early in development and are characterized by deficits in personal, social, academic, or occupational functioning. These disorders frequently co-occur and include conditions such as autism spectrum disorder (ASD) and attention-deficit/hyperactivity disorder (ADHD). Mood disorders (MDs), such as major depressive disorder and bipolar disorder, also pose significant global health challenges due to their high prevalence and substantial impact on quality of life. Emerging evidence highlights overlapping neurobiological mechanisms between NDDs and MDs, including shared genetic susceptibilities, neurotransmitter dysregulation (e.g., dopaminergic and serotonergic pathways), neuroinflammation, and hypothalamic-pituitary-adrenal (HPA) axis dysfunction. Environmental factors such as early-life adversity further exacerbate these vulnerabilities, contributing to the complexity of their clinical presentation and comorbidity. Functional neuroimaging studies reveal altered connectivity in brain regions critical for emotional regulation and executive function, such as the prefrontal cortex and amygdala, across these disorders. Despite these advances, integrative diagnostic frameworks and targeted therapeutic strategies remain underexplored, limiting effective intervention. This review synthesizes current knowledge on the shared neurobiological underpinnings of NDDs and MDs, emphasizing the need for multidisciplinary research, including genetic, pharmacological, and psychological approaches, for unified diagnosis and treatment. Addressing these intersections can improve clinical outcomes and enhance the quality of life for individuals affected by these disorders.
Collapse
Affiliation(s)
- Amanda Gollo Bertollo
- Postgraduate Program in Neuroscience, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, SC, Brazil;
| | - Camila Ferreira Puntel
- Laboratory of Physiology, Pharmacology and Psychopathology, Federal University of Fronteira Sul (UFFS), Chapecó 89815-899, SC, Brazil; (C.F.P.); (B.V.d.S.)
| | - Brunna Varela da Silva
- Laboratory of Physiology, Pharmacology and Psychopathology, Federal University of Fronteira Sul (UFFS), Chapecó 89815-899, SC, Brazil; (C.F.P.); (B.V.d.S.)
| | - Marcio Martins
- Postgraduate Program in Contemporary Cultural Studies, Federal University of Mato Grosso (UFMT), Cuiabá 78060-900, MT, Brazil;
| | - Margarete Dulce Bagatini
- Cell Culture Laboratory, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul (UFFS), Chapecó 89815-899, SC, Brazil;
| | - Zuleide Maria Ignácio
- Laboratory of Physiology, Pharmacology and Psychopathology, Federal University of Fronteira Sul (UFFS), Chapecó 89815-899, SC, Brazil; (C.F.P.); (B.V.d.S.)
| |
Collapse
|
21
|
Cording KR, Tu EM, Wang H, Agopyan-Miu AHCW, Bateup HS. Cntnap2 loss drives striatal neuron hyperexcitability and behavioral inflexibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.09.593387. [PMID: 38766169 PMCID: PMC11100810 DOI: 10.1101/2024.05.09.593387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by two major diagnostic criteria - persistent deficits in social communication and interaction, and the presence of restricted, repetitive patterns of behavior (RRBs). Evidence from both human and animal model studies of ASD suggest that alteration of striatal circuits, which mediate motor learning, action selection, and habit formation, may contribute to the manifestation of RRBs. CNTNAP2 is a syndromic ASD risk gene, and loss of function of Cntnap2 in mice is associated with RRBs. How loss of Cntnap2 impacts striatal neuron function is largely unknown. In this study, we utilized Cntnap2 -/- mice to test whether altered striatal neuron activity contributes to aberrant motor behaviors relevant to ASD. We find that Cntnap2 -/- mice exhibit increased cortical drive of direct pathway striatal projection neurons (dSPNs). This enhanced drive is likely due to increased intrinsic excitability of dSPNs, which make them more responsive to cortical inputs. We find that Cntnap2 -/- mice exhibit spontaneous repetitive behaviors, increased motor routine learning, perseveration, and cognitive inflexibility. Increased corticostriatal drive of the direct pathway may therefore contribute to the acquisition of repetitive, inflexible behaviors in Cntnap2 mice.
Collapse
Affiliation(s)
- Katherine R. Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA USA
| | - Emilie M. Tu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Hongli Wang
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA USA
| | | | - Helen S. Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| |
Collapse
|
22
|
Gonzales S, Zhao JZ, Choi NY, Acharya P, Jeong S, Wang X, Lee MY. SOX7: Autism Associated Gene Identified by Analysis of Multi-Omics Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.05.26.542456. [PMID: 37292933 PMCID: PMC10245991 DOI: 10.1101/2023.05.26.542456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Genome-wide association studies and next generation sequencing data analyses based on DNA information have identified thousands of mutations associated with autism spectrum disorder (ASD). However, more than 99% of identified mutations are non-coding. Thus, it is unclear which of these mutations might be functional and thus potentially causal variants. Transcriptomic profiling using total RNA-sequencing has been one of the most utilized approaches to link protein levels to genetic information at the molecular level. The transcriptome captures molecular genomic complexity that the DNA sequence solely does not. Some mutations alter a gene's DNA sequence but do not necessarily change expression and/or protein function. To date, few common variants reliably associated with the diagnosis status of ASD despite consistently high estimates of heritability. In addition, reliable biomarkers used to diagnose ASD or molecular mechanisms to define the severity of ASD do not exist. Therefore, it is necessary to integrate DNA and RNA testing together to identify true causal genes and propose useful biomarkers for ASD. We performed gene-based association studies with adaptive test using genome-wide association studies (GWAS) summary statistics with two large GWAS datasets (ASD 2019 data: 18,382 ASD cases and 27,969 controls [discovery data]; ASD 2017 data: 6,197 ASD cases and 7,377 controls [replication data]) which were obtained from the Psychiatric Genomics Consortium (PGC). In addition, we investigated differential expression between ASD cases and controls for genes identified in gene-based GWAS with two RNA-seq datasets (GSE211154: 20 cases and 19 controls; GSE30573: 3 cases and 3 controls). We identified 5 genes significantly associated with ASD in ASD 2019 data (KIZ-AS1, p=8.67×10-10; KIZ, p=1.16×10-9; XRN2, p=7.73×10-9; SOX7, p=2.22×10-7; LOC101929229 also known as PINX1-DT, p=2.14×10-6). Among these 5 genes, gene SOX7 (p=0.00087) and LOC101929229 (p=0.009) were replicated in ASD 2017 data. KIZ-AS1 (p=0.059) and KIZ (p=0.06) were close to the boundary of replication in ASD 2017 data. Genes SOX7 (p=0.036 in all samples; p=0.044 in white samples) indicated significant expression differences between cases and controls in the GSE211154 RNA-seq data. Furthermore, gene SOX7 was upregulated in cases than in controls in the GSE30573 RNA-seq data (p=0.0017; Benjamini-Hochberg adjusted p=0.0085). SOX7 encodes a member of the SOX (SRY-related HMG-box) family of transcription factors pivotally contributing to determining of the cell fate and identity in many lineages. The encoded protein may act as a transcriptional regulator after forming a protein complex with other proteins leading to autism. Gene SOX7 in the transcription factor family could be associated with ASD. This finding may provide new diagnostic and therapeutic strategies for ASD.
Collapse
Affiliation(s)
- Samantha Gonzales
- Department of Biostatistics, Florida International University, Miami, FL 33199
| | - Jane Zizhen Zhao
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Na Young Choi
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207
| | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207
| | - Sehoon Jeong
- Department of Healthcare Information Technology Inje University, Gimhae, South Korea, 50834
| | - Xuexia Wang
- Department of Biostatistics, Florida International University, Miami, FL 33199
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207
| |
Collapse
|
23
|
Koko M, Satterstrom FK, Warrier V, Martin H. Contribution of autosomal rare and de novo variants to sex differences in autism. Am J Hum Genet 2025; 112:599-614. [PMID: 39954678 PMCID: PMC11947420 DOI: 10.1016/j.ajhg.2025.01.016] [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: 05/07/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/17/2025] Open
Abstract
Autism is four times more prevalent in males than females. To study whether this reflects a difference in genetic predisposition attributed to autosomal rare variants, we evaluated sex differences in effect size of damaging protein-truncating and missense variants on autism predisposition in 47,061 autistic individuals using a liability model with differing thresholds. Given the sex differences in the rates of cognitive impairment among autistic individuals, we also compared effect sizes of rare variants between individuals with and without cognitive impairment or motor delay. Although these variants mediated different likelihoods of autism with versus without cognitive or motor difficulties, their effect sizes on the liability scale did not differ significantly by sex exome wide or in genes sex-differentially expressed in the cortex. De novo mutations were enriched in genes with male-biased expression in the adult cortex, but these genes did not show a significant sex difference on the liability scale, nor did the liability conferred by these genes differ significantly from other genes with similar loss-of-function intolerance and sex-averaged cortical expression. Exome-wide female bias in de novo protein-truncating mutation rates on the observed scale was driven by high-confidence and syndromic autism-predisposition genes. In summary, autosomal rare and damaging coding variants confer similar liability for autism in females and males.
Collapse
Affiliation(s)
- Mahmoud Koko
- Human Genetics, Wellcome Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK
| | - F Kyle Satterstrom
- 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; Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Varun Warrier
- Department of Psychiatry, Autism Research Centre, University of Cambridge, Cambridge, Cambridgeshire CB2 8AH, UK.
| | - Hilary Martin
- Human Genetics, Wellcome Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK.
| |
Collapse
|
24
|
Tsujimura K, Ortug A, Alatorre Warren JL, Shiohama T, McDougle CJ, Marcus RE, Tseng CEJ, Zürcher NR, Mercaldo ND, Faja S, Maunakea A, Hooker J, Takahashi E. Structural pathways related to the subventricular zone are decreased in volume with altered microstructure in young adult males with autism spectrum disorder. Cereb Cortex 2025; 35:bhaf041. [PMID: 40055911 DOI: 10.1093/cercor/bhaf041] [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: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 03/22/2025] Open
Abstract
Autism spectrum disorder is a neurodevelopmental condition characterized by reduced social communication and repetitive behaviors. Altered neurogenesis, including disturbed neuronal migration, has been implicated in autism spectrum disorder. Using diffusion MRI, we previously identified neuronal migration pathways in the human fetal brain and hypothesized that similar pathways persist into adulthood, with differences in volume and microstructural characteristics between individuals with autism spectrum disorder and controls. We analyzed diffusion MRI-based tractography of subventricular zone-related pathways in 15 young adult men with autism spectrum disorder and 18 controls at Massachusetts General Hospital, with validation through the Autism Imaging Data Exchange II dataset. Participants with autism spectrum disorder had reduced subventricular zone pathway volumes and fractional anisotropy compared to controls. Furthermore, subventricular zone pathway volume was positively correlated (r: 0.68; 95% CI: 0.25 to 0.88) with symptom severity, suggesting that individuals with more severe symptoms tended to have larger subventricular zone pathway volumes, normalized by brain size. Analysis of the Autism Imaging Data Exchange cohort confirmed these findings of reduced subventricular zone pathway volumes in autism spectrum disorder. While some of these pathways may potentially include inaccurately disconnected pathways that go through the subventricular zone, our results suggest that diffusion MRI-based tractography pathways anatomically linked to the periventricular region are associated with certain symptom types in adult males with autism spectrum disorder.
Collapse
Affiliation(s)
- Keita Tsujimura
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
- Group of Brain Function and Development, Neuroscience Institute of the Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
- Research Unit for Developmental Disorders, Institute for Advanced Research, Nagoya University, Nagoya 464-8601, Aichi, Japan
| | - Alpen Ortug
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - José Luis Alatorre Warren
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo 0317, Norway
| | - Tadashi Shiohama
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Christopher J McDougle
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA 02421, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA 02215, United States
| | - Rachel E Marcus
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA 02421, United States
| | - Chieh-En Jane Tseng
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Nicole R Zürcher
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA 02421, United States
| | - Nathaniel D Mercaldo
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Susan Faja
- Division of Developmental Medicine, Department of Pediatrics, Harvard School of Medicine, Boston, MA 02215, United States
| | - Alika Maunakea
- Department of Anatomy, Biochemistry, and Physiology (ABP), John A. Burns School of Medicine (JABSOM), University of Hawaii, Manoa, Honolulu, HI 96813, United States
| | - Jacob Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA 02421, United States
| | - Emi Takahashi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| |
Collapse
|
25
|
Mattingly Z, Chetty S. Untangling the Molecular Mechanisms Contributing to Autism Spectrum Disorder Using Stem Cells. Autism Res 2025; 18:476-485. [PMID: 39989339 DOI: 10.1002/aur.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
Autism spectrum disorder (ASD) is a complex neuro developmental condition characterized by significant genetic and phenotypic variability, making diagnosis and treatment challenging. The heterogeneity of ASD-associated genetic variants and the absence of clear causal factors in many cases complicate personalized care. Traditional models, such as postmortem brain tissue and animal studies, have provided valuable insights but are limited in capturing the dynamic processes and human-specific aspects of ASD pathology. Recent advances in human induced pluripotent stem cell (iPSC) technology have transformed ASD research by enabling the generation of patient-derived neural cells in both two-dimensional cultures and three-dimensional brain organoid models. These models retain the donor's genetic background, allowing researchers to investigate disease-specific cellular and molecular mechanisms while identifying potential therapeutic targets tailored to individual patients. This commentary highlights how stem cell-based approaches are advancing our understanding of ASD and paving the way for more personalized diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Zoe Mattingly
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sundari Chetty
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Lurie Center for Autism, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Martin H, Choi JE, Rodrigues AR, Eshel N. Review: Dopamine, Serotonin, and the Translational Neuroscience of Aggression in Autism Spectrum Disorder. JAACAP OPEN 2025; 3:29-41. [PMID: 40109493 PMCID: PMC11914923 DOI: 10.1016/j.jaacop.2024.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 03/22/2025]
Abstract
Objective Autism spectrum disorder (ASD) is a neurodevelopmental disorder with a 1% to 2% prevalence in children. In addition to social communication deficits and restricted or repetitive behavior, ASD is often characterized by a heightened propensity for aggression. In fact, aggressive behavior is the primary reason for hospitalization in children with ASD, and current treatment options, despite some efficacy, are often associated with prominent side effects. Despite such high clinical toll, the neurobiology of aggression in ASD remains poorly understood. Method The neural circuits linked to both ASD and aggression were reviewed, with the goal of identifying overlapping components to help guide future treatment development. In discussing the clinical phenotype of aggression in ASD, some of the triggers and risk factors were noted to differ from those that cause aggression in neurotypical children. Preclinical and clinical studies on the neurobiology of aggression and ASD were synthesized to combine evidence from genetics, neuroimaging, pharmacology, and circuit manipulations. Dopamine and serotonin, 2 neuromodulators that contribute to development and behavioral control, were specifically studied. Results The literature indicates that the intricate interplay of the dopamine and serotonin systems has a pivotal role in shaping behavior, including the expression of aggression. Conclusion Understanding the balance between dopamine as an accelerator and serotonin as a brake may provide insights into the mechanisms of aggression in children with ASD. Although much work remains to be done, new perspectives promise to bridge the gap between human and animal studies and pinpoint the neurobiology of aggression in ASD. Diversity & Inclusion Statement One or more of the authors of this paper self-identifies as a member of one or more historically underrepresented sexual and/or gender groups in science. We actively worked to promote sex and gender balance in our author group.
Collapse
Affiliation(s)
| | | | | | - Neir Eshel
- Stanford University, Stanford, California
| |
Collapse
|
27
|
O'Grady K, Grabrucker AM. Metal Dyshomeostasis as a Driver of Gut Pathology in Autism Spectrum Disorders. J Neurochem 2025; 169:e70041. [PMID: 40108935 PMCID: PMC11923526 DOI: 10.1111/jnc.70041] [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: 10/01/2024] [Revised: 01/31/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Despite being classified as neurodevelopmental disorders, in recent years, there has been a growing interest in the association between autism spectrum disorders (ASDs) and gut pathology. This comprehensive and systematic review explores a potential mechanism underlying gut pathology in ASDs, including alterations in gut microbiota, intestinal permeability, immune dysregulation, and gastrointestinal (GI) symptoms. Specifically, it delves into the role of toxic and essential metals and their interplay, affecting the development and function of the GI tract. The review also discusses the potential implications of this gut pathology in the development and management of ASDs. Studies have shown that heavy metal exposure, whether through environmental sources or dietary intake, can disrupt the delicate balance of trace elements in the gut. This disruption can adversely affect zinc homeostasis, potentially exacerbating gut pathology in individuals with ASDs. The impaired zinc absorption resulting from heavy metal exposure may contribute to the immune dysregulation, oxidative stress, and inflammation observed in the gut of individuals with ASDs. By shedding light on the multifaceted nature of gut pathology, including the impact of metal dyshomeostasis as a non-genetic factor in ASD, this review underscores the significance of the gut-brain axis in the etiology and management of ASDs.
Collapse
Affiliation(s)
- Katelyn O'Grady
- Department of Biological SciencesUniversity of LimerickLimerickIreland
- Bernal InstituteUniversity of LimerickLimerickIreland
- Health Research Institute (HRI)University of LimerickLimerickIreland
| | - Andreas M. Grabrucker
- Department of Biological SciencesUniversity of LimerickLimerickIreland
- Bernal InstituteUniversity of LimerickLimerickIreland
- Health Research Institute (HRI)University of LimerickLimerickIreland
| |
Collapse
|
28
|
DeSpenza T, Kiziltug E, Allington G, Barson DG, McGee S, O'Connor D, Robert SM, Mekbib KY, Nanda P, Greenberg ABW, Singh A, Duy PQ, Mandino F, Zhao S, Lynn A, Reeves BC, Marlier A, Getz SA, Nelson-Williams C, Shimelis H, Walsh LK, Zhang J, Wang W, Prina ML, OuYang A, Abdulkareem AF, Smith H, Shohfi J, Mehta NH, Dennis E, Reduron LR, Hong J, Butler W, Carter BS, Deniz E, Lake EMR, Constable RT, Sahin M, Srivastava S, Winden K, Hoffman EJ, Carlson M, Gunel M, Lifton RP, Alper SL, Jin SC, Crair MC, Moreno-De-Luca A, Luikart BW, Kahle KT. PTEN mutations impair CSF dynamics and cortical networks by dysregulating periventricular neural progenitors. Nat Neurosci 2025; 28:536-557. [PMID: 39994410 PMCID: PMC12038823 DOI: 10.1038/s41593-024-01865-3] [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: 07/06/2023] [Accepted: 12/05/2024] [Indexed: 02/26/2025]
Abstract
Enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles (ventriculomegaly) is a defining feature of congenital hydrocephalus (CH) and an under-recognized concomitant of autism. Here, we show that de novo mutations in the autism risk gene PTEN are among the most frequent monogenic causes of CH and primary ventriculomegaly. Mouse Pten-mutant ventriculomegaly results from aqueductal stenosis due to hyperproliferation of periventricular Nkx2.1+ neural progenitor cells (NPCs) and increased CSF production from hyperplastic choroid plexus. Pten-mutant ventriculomegalic cortices exhibit network dysfunction from increased activity of Nkx2.1+ NPC-derived inhibitory interneurons. Raptor deletion or postnatal everolimus treatment corrects ventriculomegaly, rescues cortical deficits and increases survival by antagonizing mTORC1-dependent Nkx2.1+ NPC pathology. Thus, PTEN mutations concurrently alter CSF dynamics and cortical networks by dysregulating Nkx2.1+ NPCs. These results implicate a nonsurgical treatment for CH, demonstrate a genetic association of ventriculomegaly and ASD, and help explain neurodevelopmental phenotypes refractory to CSF shunting in select individuals with CH.
Collapse
Affiliation(s)
- Tyrone DeSpenza
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Emre Kiziltug
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Garrett Allington
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons and New York Presbyterian Hospital, New York, NY, USA
| | - Daniel G Barson
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | | | - David O'Connor
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie M Robert
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Kedous Y Mekbib
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Pranav Nanda
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ana B W Greenberg
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Amrita Singh
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Phan Q Duy
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Francesca Mandino
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Shujuan Zhao
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Anna Lynn
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Benjamin C Reeves
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Arnaud Marlier
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Stephanie A Getz
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Carol Nelson-Williams
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Hermela Shimelis
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
| | - Lauren K Walsh
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
| | - Junhui Zhang
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Wei Wang
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Mackenzi L Prina
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA
| | - Annaliese OuYang
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Asan F Abdulkareem
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA
| | - Hannah Smith
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - John Shohfi
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Neel H Mehta
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Evan Dennis
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laetitia R Reduron
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Jennifer Hong
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - William Butler
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Engin Deniz
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Evelyn M R Lake
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - R Todd Constable
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Siddharth Srivastava
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kellen Winden
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ellen J Hoffman
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Marina Carlson
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Seth L Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Diagnostic Medicine Institute, Geisinger, Danville, PA, USA
| | - Sheng Chih Jin
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael C Crair
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Andres Moreno-De-Luca
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
- Department of Radiology, Diagnostic Medicine Institute, Geisinger, Danville, PA, USA
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA.
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA.
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
29
|
Blázquez A, Rodriguez-Revenga L, Alvarez-Mora MI, Calvo R. Clinical and genetic findings in autism spectrum disorders analyzed using exome sequencing. Front Psychiatry 2025; 16:1515793. [PMID: 40071278 PMCID: PMC11893851 DOI: 10.3389/fpsyt.2025.1515793] [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: 10/23/2024] [Accepted: 01/30/2025] [Indexed: 03/14/2025] Open
Abstract
Autism spectrum disorder (ASD) refers to a group of complex neurodevelopmental disorders and is characterized by impaired reciprocal social interaction and communication, as well as the presence of restricted interests and stereotyped and repetitive behaviors. As a complex neurodevelopmental disorder, the phenotype and severity of autism are extremely heterogeneous, with differences from one patient to another. Chromosome microarray (CMA) and fragile X syndrome analyses has been used as a powerful tool to identify new candidate genes for ASD. METHODS In the present study, CMA was first used to scan for genome-wide copy number variants in the patient, and no clinically significant copy number variants were found. Exome sequencing (ES) was used for further genetic testing. RESULTS ES was performed on 20 subjects. Eighty percent of our sample presented intellectual disability. Other co-occurring clinical conditions included speech disorders, psychomotor delay, the presence of dysmorphic features and medical co-morbidities. A pathogenic variant was identified in 10 patients (ADNP, FBN1, WAC, ASXL3, NR4A2, ALX4, ANKRD1, POGZ, SHANK3 and BPTF). Patients with a positive finding in ES were more likely to present a dysmorphic trunk, more than three dysmorphic features, hypotonia, psychomotor delay and strabismus. CONCLUSIONS ES offers expanded diagnostic options for patients with ASD who are negative on CMA. However, further studies are needed for a better understanding of ASD etiology and also the different phenotypes.
Collapse
Affiliation(s)
- Ana Blázquez
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neuroscience, Hospital Clínic de Barcelona, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Basic Clinal Practice, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Laia Rodriguez-Revenga
- Department of Biochemistry and Molecular Genetics, Hospital Clinic of Barcelona, Barcelona, Spain
- Translational Research Group in New Therapeutic and Diagnostic Strategies in Liver Diseases Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - María I. Alvarez-Mora
- Department of Biochemistry and Molecular Genetics, Hospital Clinic of Barcelona, Barcelona, Spain
- Translational Research Group in New Therapeutic and Diagnostic Strategies in Liver Diseases Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Calvo
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neuroscience, Hospital Clínic de Barcelona, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
30
|
Qureshi AY, Nielsen JA, Sepulcre J. Differential links in 16p11.2 deletion carriers reveal aberrant connections between large-scale networks. Cereb Cortex 2025; 35:bhae474. [PMID: 40007052 PMCID: PMC11859958 DOI: 10.1093/cercor/bhae474] [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: 06/25/2024] [Revised: 10/25/2024] [Accepted: 12/02/2024] [Indexed: 02/27/2025] Open
Abstract
Qualitatively different topographical patterns of connections are thought to underlie individual differences in thought and behavior, particularly at heteromodal association areas. As such, we hypothesized that connections unique to 16p11.2 deletion carriers compared to controls, rather than hyper- or hypo-connectivity, would serve as a better model to explain the cognitive and behavioral changes observed in individuals carrying this autism-risk copy number variation. Using a spatially-unbiased, data-driven approach we found that differential links clustered non-uniformly across the cortex-particularly at the superior temporal gyrus and sulcus, posterior insula, cingulate sulcus, and inferior parietal lobule bilaterally. At these hotspots, altered local connectivity that spanned across the borders of cortical large-scale networks coincided with aberrant distant interconnectivity between large-scale networks. This was most evident between the auditory and the dorsomedial default (DNb) networks-such that greater between-network interconnectivity was associated with greater communication and social impairment. Entangled connectivity between large-scale networks may preclude each network from having the necessary fidelity to operate properly, particularly when the 2 networks have opposing organization principles-namely, local specialization (segregation) versus global coherency (integration).
Collapse
Affiliation(s)
- Abid Y Qureshi
- Department of Neurology, University of Kansas Medical Center, 2100 W 36th Ave, Suite 130J, Kansas City, KS 66103, United States
| | - Jared A Nielsen
- Department of Psychology and Neuroscience Center, KMBL 1001, Brigham Young University, Provo, UT 84602, United States
| | - Jorge Sepulcre
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, 330 Cedar St, New Haven, CT 06520, United States
- Gordon Center for Medical Imaging, Department of Radiology, Mass General Brigham, 125 Nashua Street, Boston, MA 02114, United States
| |
Collapse
|
31
|
Hsueh YP. Signaling in autism: Relevance to nutrients and sex. Curr Opin Neurobiol 2025; 90:102962. [PMID: 39731919 DOI: 10.1016/j.conb.2024.102962] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 11/11/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024]
Abstract
Autism spectrum disorders (ASD) are substantially heterogeneous neuropsychiatric conditions with over a thousand associated genetic factors and various environmental influences, such as infection and nutrition. Additionally, males are four times more likely than females to be affected. This heterogeneity underscores the need to uncover common molecular features within ASD. Recent studies have revealed interactions among genetic predispositions, environmental factors, and sex that may be critical to ASD etiology. This review focuses on emerging evidence for the impact of nutrients-particularly zinc and amino acids-on ASD, as demonstrated in mouse models and human studies. These nutrients have been shown to influence synaptic signaling, dendritic spine formation, and behaviors linked to autism. Furthermore, sex-based differences in nutritional requirements, especially for zinc and amino acids, may contribute to the observed male bias in autism, indicating that interactions between nutrients and genetic factors could be integral to understanding and potentially mitigating ASD symptoms.
Collapse
Affiliation(s)
- Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan, ROC.
| |
Collapse
|
32
|
Mahajan P, Patil D, Nair N, Musmade N, Apte P. Mapping the Landscape of Autism Research: A Scientometric Review (2011-2023). Int J Dev Neurosci 2025; 85:e10406. [PMID: 39723621 DOI: 10.1002/jdn.10406] [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: 09/10/2024] [Revised: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
This scientometric analysis maps the landscape of autism spectrum disorder (ASD) research between 2011 and 2023. By exploring patterns in publication growth, geographic distribution and institutional involvement, this study highlights evolving research themes, key contributors and collaborative networks. Our findings reveal a marked rise in ASD publications, particularly from 2020 onwards, with the United States, United Kingdom and China leading in contributions and collaborations. Scientometric analysis identifies a shift towards advanced machine learning techniques and neuroimaging in ASD studies, reflecting technological integration in research. Institutional analysis uncovers Vanderbilt University and Yale University as major contributors, with significant citation impacts across their publications. Furthermore, prominent funding sources, including the National Institutes of Health, underscore the critical role of funding in shaping research priorities. This comprehensive scientometric overview not only consolidates current knowledge but also serves as a resource to inform future research directions, enhancing interdisciplinary approaches to ASD understanding and intervention.
Collapse
Affiliation(s)
- Pratibha Mahajan
- Department of Artificial Intelligence, Vishwakarma University, Pune, India
| | - Deven Patil
- Department of Artificial Intelligence, Vishwakarma University, Pune, India
| | - Nidhi Nair
- Department of Artificial Intelligence, Vishwakarma University, Pune, India
| | - Nishant Musmade
- Department of Artificial Intelligence, Vishwakarma University, Pune, India
| | - Preet Apte
- Department of Artificial Intelligence, Vishwakarma University, Pune, India
| |
Collapse
|
33
|
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
|
34
|
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
|
35
|
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
|
36
|
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
|
37
|
Hurtado EC, Wotton JM, Gulka A, Burke C, Ng JK, Bah I, Manuel J, Heins H, Murray SA, Gorkin DU, White JK, Peterson KA, Turner TN. Generation and Characterization of a Knockout Mouse of an Enhancer of EBF3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.631762. [PMID: 39829799 PMCID: PMC11741297 DOI: 10.1101/2025.01.09.631762] [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
Genomic studies of autism and other neurodevelopmental disorders have identified several relevant protein-coding and noncoding variants. One gene with an excess of protein-coding de novo variants is EBF3 that also is the gene underlying the Hypotonia, Ataxia, and Delayed Development Syndrome (HADDS). In previous work, we have identified noncoding de novo variants in an enhancer of EBF3 called hs737 and further showed that there was an enrichment of deletions of this enhancer in individuals with neurodevelopmental disorders. In this present study, we generated a novel mouse line that deletes the highly conserved, orthologous mouse region of hs737 within the Rr169617 regulatory region, and characterized the molecular and phenotypic aspects of this mouse model. This line contains a 1,160 bp deletion within Rr169617 and through heterozygous crosses we found a deviation from Mendelian expectation (p = 0.02) with a significant depletion of the deletion allele (p = 5.8 × 10-4). Rr169617 +/- mice had a reduction of Ebf3 expression by 10% and Rr169617 -/- mice had a reduction of Ebf3 expression by 20%. Differential expression analyses in E12.5 forebrain, midbrain, and hindbrain in Rr169617 +/+ versus Rr169617 -/- mice identified dysregulated genes including histone genes (i.e., Hist1h1e, Hist1h2bk, Hist1h3i, Hist1h2ao) and other brain development related genes (e.g., Chd5, Ntng1). A priori phenotyping analysis (open field, hole board and light/dark transition) identified sex-specific differences in behavioral traits when comparing Rr169617 -/- males versus females; whereby, males were observed to be less mobile, move slower, and spend more time in the dark. Furthermore, both sexes when homozygous for the enhancer deletion displayed body composition differences when compared to wild-type mice. Overall, we show that deletion within Rr169617 reduces the expression of Ebf3 and results in phenotypic outcomes consistent with potential sex specific behavioral differences. This enhancer deletion line provides a valuable resource for others interested in noncoding regions in neurodevelopmental disorders and/or those interested in the gene regulatory network downstream of Ebf3.
Collapse
Affiliation(s)
- Emily Cordova Hurtado
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Alexander Gulka
- Department of Biology, Emory University. Atlanta, GA 30322, USA
| | | | - Jeffrey K. Ng
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ibrahim Bah
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Juana Manuel
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hillary Heins
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - David U. Gorkin
- Department of Biology, Emory University. Atlanta, GA 30322, USA
| | | | | | - Tychele N. Turner
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
38
|
Chavez J, Wolf T, Geng Z, Tai YT, Bright K, Stafford J, Gao Z. The zinc-finger protein POGZ associates with Polycomb repressive complex 1 to regulate bone morphogenetic protein signaling during neuronal differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631780. [PMID: 39829763 PMCID: PMC11741289 DOI: 10.1101/2025.01.07.631780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Polycomb Repressive Complex 1 (PRC1) is a family of epigenetic regulators critical for mammalian development. Elucidating PRC1 composition and function across cell types and developmental stages is key to understanding the epigenetic regulation of cell fate determination. In this study, we discovered POGZ, a prominent Autism Spectrum Disorder (ASD) risk factor, as a novel component of PRC1.6, forming the PRC1.6-POGZ complex. Functional assays revealed that POGZ elicits transcriptional repression that is dependent on RING1B expression. Analysis of publicly available ChIP-Seq data showed that POGZ highly colocalizes with RING1B and HP1γ, two PRC1.6 components, at genes involved in multiple aspects of transcriptional regulation in the embryonic mouse cortex. Although Pogz knockout (KO) does not compromise stem cell pluripotency, Pogz ablation in neuronal progenitor cells (NPCs) led to widespread transcriptomic dysregulation with failed activation of key neuronal genes. Finally, we demonstrated that PRC1.6-POGZ regulates neuronal differentiation by repressing the bone morphogenetic protein (BMP) signaling pathway. These findings reveal a mechanism by which PRC1 and POGZ coordinate transcriptional regulation during neuronal differentiation, which offers insights into how disruptions in this pathway may contribute to neurodevelopmental disorders such as ASD.
Collapse
|
39
|
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
|
40
|
Tien IS, Johnson AR, Kim J, Wood JJ. Examining Diagnostic Trends and Gender Differences in the ADOS-II. J Autism Dev Disord 2025; 55:1-9. [PMID: 38064005 DOI: 10.1007/s10803-023-06191-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2023] [Indexed: 02/07/2025]
Abstract
Approximately 3-4 boys for every girl meet the clinical criteria for autism in studies of community diagnostic patterns and studies of autism using samples of convenience. However, girls with autism have been hypothesized to be underdiagnosed, possibly because they may present with differing symptom profiles as compared to boys. This secondary data analysis used the National Database of Autism Research (NDAR) to examine how gender and symptom profiles are associated with one another in a gold standard assessment of autism symptoms, the Autism Diagnostic Observation Schedule II (ADOS-II; Lord, C., Luyster, R., Guthrie, W., & Pickles A. (2012a). Patterns of developmental trajectories in toddlers with autism spectrum disorder. Journal of Consulting and Clinical Psychology, 80(3):477-489. https://doi.org/10.1037/a0027214 . Epub 2012 Apr 16. PMID: 22506796, PMCID: PMC3365612). ADOS-II scores from 6183 children ages 6-14 years from 78 different studies in the NDAR indicated that gender was a significant predictor of total algorithm, restrictive and repetitive behavioral, and social communicative difficulties composite severity scores. These findings suggest that gender differences in ADOS scores are common in many samples and may reflect on current diagnostic practices.
Collapse
Affiliation(s)
- Ingrid S Tien
- University of California, Los Angeles Department of Education, 300 Charles E. Young Blvd, Room 208A, 90095, Los Angeles, CA, USA.
| | - Amanda R Johnson
- University of California, Los Angeles Department of Education, 300 Charles E. Young Blvd, Room 208A, 90095, Los Angeles, CA, USA
| | - Junok Kim
- University of California, Los Angeles Department of Education, 300 Charles E. Young Blvd, Room 208A, 90095, Los Angeles, CA, USA
| | - Jeffrey J Wood
- University of California, Los Angeles Department of Education, 300 Charles E. Young Blvd, Room 208A, 90095, Los Angeles, CA, USA
| |
Collapse
|
41
|
Singh NK, Patel A, Verma N, Singh RKB, Sharma SK. Hybrid deep learning method to identify key genes in autism spectrum disorder. Healthc Technol Lett 2025; 12:e12104. [PMID: 40292027 PMCID: PMC12023764 DOI: 10.1049/htl2.12104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 04/30/2025] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with a strong genetic component. This research aims to identify key genes associated with autism spectrum disorder using a hybrid deep learning approach. To achieve this, a protein-protein interaction network is constructedand analyzed through a graph convolutional network, which extracts features based on gene interactions. Logistic regression is then employed to predict potential key regulatorgenes using probability scores derived from these features. To evaluate the infection ability of these potential key regulator genes, a susceptible-infected (SI) model, is performed, which reveals the higher infection ability for the genes identified by the proposed method, highlighting its effectiveness in pinpointing key genetic factors associated with ASD. The performance of the proposed method is compared with centrality methods, showing significantly improved results. Identified key genes are further compared with the SFARI gene database and the Evaluation of Autism Gene Link Evidence (EAGLE) framework, revealing commongenes that are strongly associated with ASD. This reinforces the validity of the method in identifying key regulator genes. The proposed method aligns with advancements in therapeutic systems, diagnostics, and neural engineering, providing a robust framework for ASD research and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Naveen Kumar Singh
- School of Computer and Systems SciencesJawaharlal Nehru UniversityNew DelhiIndia
| | - Asmita Patel
- School of Computer and Systems SciencesJawaharlal Nehru UniversityNew DelhiIndia
| | - Nidhi Verma
- Department of MicrobiologyRam Lal Anand CollegeUniversity of DelhiNew DelhiIndia
| | - R. K. Brojen Singh
- School of Computational and Integrative SciencesJawaharlal Nehru UniversityNew DelhiIndia
| | - Saurabh Kumar Sharma
- School of Computer and Systems SciencesJawaharlal Nehru UniversityNew DelhiIndia
| |
Collapse
|
42
|
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
|
43
|
Bo M, Carta A, Cipriani C, Cavassa V, Simula ER, Huyen NT, Phan GTH, Noli M, Matteucci C, Sotgiu S, Balestrieri E, Sechi LA. HERVs Endophenotype in Autism Spectrum Disorder: Human Endogenous Retroviruses, Specific Immunoreactivity, and Disease Association in Different Family Members. Microorganisms 2024; 13:9. [PMID: 39858776 PMCID: PMC11767913 DOI: 10.3390/microorganisms13010009] [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: 11/27/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Increasing evidence indicates that human endogenous retroviruses (HERVs) are important to human health and are an underexplored component of many diseases. Certain HERV families show unique expression patterns and immune responses in autism spectrum disorder (ASD) patients compared to healthy controls, suggesting their potential as biomarkers. Despite these interesting findings, the role of HERVs in ASD needs to be further investigated. In this review, we discuss recent advances in genetic research on ASD, with a particular emphasis on the implications of HERVs on neurodevelopment and future genomic initiatives aimed at discovering ASD-related genes through Artificial Intelligence. Given their pro-inflammatory and autoimmune characteristics, the existing literature suggests that HERVs may contribute to the onset or worsening of ASD in individuals with a genetic predisposition. Therefore, we propose that investigating their fundamental properties could not only improve existing therapies but also pave the way for new therapeutic strategies.
Collapse
Affiliation(s)
- Marco Bo
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Sassari, Viale San Pietro 43b, 07100 Sassari, Italy; (M.B.); (E.R.S.); (M.N.); (L.A.S.)
- Struttura Complessa Microbiologia e Virologia, Azienda Ospedaliera Universitaria Sassari, 07100 Sassari, Italy
| | - Alessandra Carta
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.)
| | - Chiara Cipriani
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (C.M.)
| | - Vanna Cavassa
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.)
| | - Elena Rita Simula
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Sassari, Viale San Pietro 43b, 07100 Sassari, Italy; (M.B.); (E.R.S.); (M.N.); (L.A.S.)
| | - Nguyen Thi Huyen
- Department of Immunology and Pathophysiology, Hue University of Medicine and Pharmacy, Hue City 53000, Vietnam; (N.T.H.); (G.T.H.P.)
| | - Giang Thi Hang Phan
- Department of Immunology and Pathophysiology, Hue University of Medicine and Pharmacy, Hue City 53000, Vietnam; (N.T.H.); (G.T.H.P.)
| | - Marta Noli
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Sassari, Viale San Pietro 43b, 07100 Sassari, Italy; (M.B.); (E.R.S.); (M.N.); (L.A.S.)
- Struttura Complessa Microbiologia e Virologia, Azienda Ospedaliera Universitaria Sassari, 07100 Sassari, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (C.M.)
| | - Stefano Sotgiu
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.)
| | - Emanuela Balestrieri
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (C.M.)
| | - Leonardo Antonio Sechi
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Sassari, Viale San Pietro 43b, 07100 Sassari, Italy; (M.B.); (E.R.S.); (M.N.); (L.A.S.)
- Struttura Complessa Microbiologia e Virologia, Azienda Ospedaliera Universitaria Sassari, 07100 Sassari, Italy
| |
Collapse
|
44
|
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
|
45
|
Kushima I, Nakatochi M, Ozaki N. Copy Number Variations and Human Well-Being: Integrating Psychiatric, Physical, and Socioeconomic Perspectives. Biol Psychiatry 2024:S0006-3223(24)01788-8. [PMID: 39643102 DOI: 10.1016/j.biopsych.2024.11.019] [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: 04/16/2024] [Revised: 11/12/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024]
Abstract
Copy number variations (CNVs) have emerged as crucial genetic factors that influence a wide spectrum of human health outcomes, with particularly strong associations to psychiatric disorders. In this review, we present a synthesis of diverse impacts of psychiatric disorder-associated CNVs on neurodevelopment, brain function, and physical health across the lifespan. Large-scale studies have revealed that CNV carriers exhibit an increased risk for psychiatric disorders, cognitive deficits, sleep disturbances, neurological disorders, and other physical conditions, including cardiovascular diseases, diabetes, and renal disease, highlighting the wide-ranging impact of CNVs beyond the brain. Neuroimaging studies have revealed substantial CNV effects on brain structure, from cortical and subcortical alterations to white matter microstructure, with effect sizes often exceeding those observed in idiopathic psychiatric disorders. Cellular and animal models have begun to elucidate dynamic CNV effects on neurodevelopment, neuronal function, and cellular energy metabolism, while revealing complex CNV-environment interactions and cell type-specific responses, particularly in studies of 22q11.2 deletion syndrome. This review also explores the complex interplay between psychiatric and physical health conditions in CNV carriers and how these interactions contribute to adverse socioeconomic outcomes, including reduced educational attainment and income levels, creating a feedback loop that further impacts health outcomes. Finally, in this review, we also highlight research limitations and propose key priorities for clinical implementation, including the need for longitudinal studies, standardized guidelines for CNV result reporting and genetic counseling, and integrated care networks to provide a foundation for advancing the field of precision psychiatry.
Collapse
Affiliation(s)
- Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan.
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Japan; Institute for Glyco-core Research, Nagoya University, Nagoya, Japan
| |
Collapse
|
46
|
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
|
47
|
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
|
48
|
Manghi P, Filosi M, Zolfo M, Casten LG, Garcia-Valiente A, Mattevi S, Heidrich V, Golzato D, Perini S, Thomas AM, Montalbano S, Cancellieri S, Waldron L, Hall JB, Xu S, Volfovsky N, Green Snyder L, Feliciano P, Asnicar F, Valles-Colomer M, Michaelson JJ, Segata N, Domenici E. Large-scale metagenomic analysis of oral microbiomes reveals markers for autism spectrum disorders. Nat Commun 2024; 15:9743. [PMID: 39528484 PMCID: PMC11555315 DOI: 10.1038/s41467-024-53934-7] [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: 04/13/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
The link between the oral microbiome and neurodevelopmental disorders remains a compelling hypothesis, still requiring confirmation in large-scale datasets. Leveraging over 7000 whole-genome sequenced salivary samples from 2025 US families with children diagnosed with autism spectrum disorders (ASD), our cross-sectional study shows that the oral microbiome composition can discriminate ASD subjects from neurotypical siblings (NTs, AUC = 0.66), with 108 differentiating species (q < 0.005). The relative abundance of these species is highly correlated with cognitive impairment as measured by Full-Scale Intelligence Quotient (IQ). ASD children with IQ < 70 also exhibit lower microbiome strain sharing with parents (p < 10-6) with respect to NTs. A two-pronged functional enrichment analysis suggests the contribution of enzymes from the serotonin, GABA, and dopamine degradation pathways to the distinct microbial community compositions observed between ASD and NT samples. Although measures of restrictive eating diet and proxies of oral hygiene show relatively minor effects on the microbiome composition, the observed associations with ASD and IQ may still represent unaccounted-for underlying differences in lifestyle among groups. While causal relationships could not be established, our study provides substantial support to the investigation of oral microbiome biomarkers in ASD.
Collapse
Affiliation(s)
- Paolo Manghi
- Department CIBIO, University of Trento, Trento, Italy.
- Computational Biology Unit, Research and Innovation Centre, Fondazione Edmund Mach, via Mach 1, 38098, San Michele all'Adige, Italy.
| | - Michele Filosi
- Department CIBIO, University of Trento, Trento, Italy
- EURAC Research Institute for Biomedicine BIO, Bolzano, Italy
| | - Moreno Zolfo
- Department CIBIO, University of Trento, Trento, Italy
- Okinawa Institute of Science and Technology (OIST), Okinawa, Japan
| | - Lucas G Casten
- Department of Psychiatry, University of Iowa, Iowa city, IA, USA
| | | | - Stefania Mattevi
- Department CIBIO, University of Trento, Trento, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | | | - Samuel Perini
- Department CIBIO, University of Trento, Trento, Italy
| | | | - Simone Montalbano
- Department CIBIO, University of Trento, Trento, Italy
- Institute of Biological Psychiatry, Copenhagen University Hospital, Copenhagen, Denmark
| | - Samuele Cancellieri
- Department CIBIO, University of Trento, Trento, Italy
- Norwegian Center of Molecular Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Levi Waldron
- CUNY Graduate School of Public Health and Health Policy, Institute for Implementation Science in Public Health, New York, NY, USA
| | | | - Simon Xu
- Simons Foundation, New York, NY, USA
| | | | - LeeAnne Green Snyder
- Simons Foundation, New York, NY, USA
- Department of Pediatrics, Division of Genetics & Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Pamela Feliciano
- Simons Foundation, New York, NY, USA
- Department of Pediatrics, Division of Genetics & Genomics, Boston Children's Hospital, Boston, MA, USA
| | | | | | | | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy.
- IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | | |
Collapse
|
49
|
Bajikar SS, Sztainberg Y, Trostle AJ, Tirumala HP, Wan YW, Harrop CL, Bengtsson JD, Carvalho CMB, Pehlivan D, Suter B, Neul JL, Liu Z, Jafar-Nejad P, Rigo F, Zoghbi HY. Modeling antisense oligonucleotide therapy in MECP2 duplication syndrome human iPSC-derived neurons reveals gene expression programs responsive to MeCP2 levels. Hum Mol Genet 2024; 33:1986-2001. [PMID: 39277796 PMCID: PMC11555823 DOI: 10.1093/hmg/ddae135] [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: 11/01/2023] [Revised: 08/12/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024] Open
Abstract
Genomic copy-number variations (CNVs) that can cause neurodevelopmental disorders often encompass many genes, which complicates our understanding of how individual genes within a CNV contribute to pathology. MECP2 duplication syndrome (MDS or MRXSL in OMIM; OMIM#300260) is one such CNV disorder caused by duplications spanning methyl CpG-binding protein 2 (MECP2) and other genes on Xq28. Using an antisense oligonucleotide (ASO) to normalize MECP2 dosage is sufficient to rescue abnormal neurological phenotypes in mouse models overexpressing MECP2 alone, implicating the importance of increased MECP2 dosage within CNVs of Xq28. However, because MDS CNVs span MECP2 and additional genes, we generated human neurons from multiple MDS patient-derived induced pluripotent cells (iPSCs) to evaluate the benefit of using an ASO against MECP2 in a MDS human neuronal context. Importantly, we identified a signature of genes that is partially and qualitatively modulated upon ASO treatment, pinpointed genes sensitive to MeCP2 function, and altered in a model of Rett syndrome, a neurological disorder caused by loss of MeCP2 function. Furthermore, the signature contained genes that are aberrantly altered in unaffected control human neurons upon MeCP2 depletion, revealing gene expression programs qualitatively sensitive to MeCP2 levels in human neurons. Lastly, ASO treatment led to a partial rescue of abnormal neuronal morphology in MDS neurons. All together, these data demonstrate that ASOs targeting MECP2 benefit human MDS neurons. Moreover, our study establishes a paradigm by which to evaluate the contribution of individual genes within a CNV to pathogenesis and to assess their potential as a therapeutic target.
Collapse
Affiliation(s)
- Sameer S Bajikar
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Cell Biology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22903, United States
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Charlottesville, VA 22903, United States
| | - Yehezkel Sztainberg
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
| | - Alexander J Trostle
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Harini P Tirumala
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
| | - Ying-Wooi Wan
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
| | - Caroline L Harrop
- Department of Cell Biology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22903, United States
| | - Jesse D Bengtsson
- Pacific Northwest Research Institute, 720 Broadway, Seattle, WA 98122, United States
| | - Claudia M B Carvalho
- Pacific Northwest Research Institute, 720 Broadway, Seattle, WA 98122, United States
| | - Davut Pehlivan
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Section of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Texas Children’s Hospital, 6621 Fannin Street, Houston, TX 77030, United States
| | - Bernhard Suter
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Section of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Texas Children’s Hospital, 6621 Fannin Street, Houston, TX 77030, United States
| | - Jeffrey L Neul
- Vanderbilt Kennedy Center, 110 Magnolia Circle, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Paymaan Jafar-Nejad
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, United States
| | - Frank Rigo
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, United States
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Section of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Texas Children’s Hospital, 6621 Fannin Street, Houston, TX 77030, United States
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, United States
| |
Collapse
|
50
|
Auwerx C, Kutalik Z, Reymond A. The pleiotropic spectrum of proximal 16p11.2 CNVs. Am J Hum Genet 2024; 111:2309-2346. [PMID: 39332410 PMCID: PMC11568765 DOI: 10.1016/j.ajhg.2024.08.015] [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: 03/28/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/29/2024] Open
Abstract
Recurrent genomic rearrangements at 16p11.2 BP4-5 represent one of the most common causes of genomic disorders. Originally associated with increased risk for autism spectrum disorder, schizophrenia, and intellectual disability, as well as adiposity and head circumference, these CNVs have since been associated with a plethora of phenotypic alterations, albeit with high variability in expressivity and incomplete penetrance. Here, we comprehensively review the pleiotropy associated with 16p11.2 BP4-5 rearrangements to shine light on its full phenotypic spectrum. Illustrating this phenotypic heterogeneity, we expose many parallels between findings gathered from clinical versus population-based cohorts, which often point to the same physiological systems, and emphasize the role of the CNV beyond neuropsychiatric and anthropometric traits. Revealing the complex and variable clinical manifestations of this CNV is crucial for accurate diagnosis and personalized treatment strategies for carrier individuals. Furthermore, we discuss areas of research that will be key to identifying factors contributing to phenotypic heterogeneity and gaining mechanistic insights into the molecular pathways underlying observed associations, while demonstrating how diversity in affected individuals, cohorts, experimental models, and analytical approaches can catalyze discoveries.
Collapse
Affiliation(s)
- Chiara Auwerx
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland; Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Zoltán Kutalik
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|