1
|
Barelle PY, Sicardi A, Schaller F, Buron J, Becquet D, Omnes F, Watrin F, Alifrangis MS, Santos C, Menuet C, François-Bellan AM, Caron E, Klucznik J, Prevot V, Bouret SG, Muscatelli F. Investigation of a mouse model of Prader-Willi Syndrome with combined disruption of Necdin and Magel2. JCI Insight 2025; 10:e185159. [PMID: 40048253 PMCID: PMC12016932 DOI: 10.1172/jci.insight.185159] [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: 04/23/2025] Open
Abstract
Prader-Willi syndrome (PWS) is a multigenic disorder caused by the loss of 7 contiguous paternally expressed genes. Mouse models with inactivation of all PWS genes are lethal. KO mouse models for each candidate gene have been generated, but they lack the functional interactions between PWS genes. Here, we revealed an interplay between Necdin and Magel2 PWS genes and generated a mouse model (named Del Ndn-Magel2 mice) with a deletion including both genes. A subset of Del Ndn-Magel2 mice showed neonatal lethality. Behaviorally, surviving mutant mice exhibited sensory delays during infancy and alterations in social exploration at adulthood. Del Ndn-Magel2 mice had a lower body weight before weaning, persisting after weaning in males only, with reduced fat mass and improved glucose tolerance as well as altered puberty. Adult mutant mice displayed increased ventilation and a persistent increase in apneas following a hypercapnic challenge. Transcriptomics analyses revealed a dysregulation of key circadian genes and alterations of genes associated with axonal function similar to patients with PWS. At neuroanatomical levels, Del Ndn-Magel2 mice had an impaired maturation of oxytocin neurons and a disrupted development of melanocortin circuits. Together, these data indicate that the Del Ndn-Magel2 mouse is a pertinent and genetically relevant model of PWS.
Collapse
Affiliation(s)
- Pierre-Yves Barelle
- University Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France
- FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Alicia Sicardi
- University Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France
- FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Fabienne Schaller
- Institut de Neurobiologie de la Méditerranée (INMED), INSERM, Aix Marseille Université, Marseille, France
| | - Julie Buron
- Institut de Neurobiologie de la Méditerranée (INMED), INSERM, Aix Marseille Université, Marseille, France
| | - Denis Becquet
- University of Aix-Marseille, Inst Neurophysiopathol, Marseille, France
| | - Felix Omnes
- Institut de Neurobiologie de la Méditerranée (INMED), INSERM, Aix Marseille Université, Marseille, France
| | - Françoise Watrin
- Institut de Neurobiologie de la Méditerranée (INMED), INSERM, Aix Marseille Université, Marseille, France
| | - Marie-Sophie Alifrangis
- Institut de Neurobiologie de la Méditerranée (INMED), INSERM, Aix Marseille Université, Marseille, France
| | | | - Clément Menuet
- Institut de Neurobiologie de la Méditerranée (INMED), INSERM, Aix Marseille Université, Marseille, France
| | | | - Emilie Caron
- University Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France
- FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Jessica Klucznik
- University Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France
- FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Vincent Prevot
- University Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France
- FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Sebastien G. Bouret
- University Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France
- FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Françoise Muscatelli
- Institut de Neurobiologie de la Méditerranée (INMED), INSERM, Aix Marseille Université, Marseille, France
| |
Collapse
|
2
|
Tamada K, Takumi T. Neurodevelopmental impact of CNV models in ASD: Recent advances and future directions. Curr Opin Neurobiol 2025; 92:103001. [PMID: 40090136 DOI: 10.1016/j.conb.2025.103001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/18/2025]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social communication impairments and restricted, repetitive behaviors. ASD exhibits a strong genetic basis, with rare and common genetic variants contributing to its etiology. Copy number variations (CNVs), deletions or duplications of chromosomal segments, have emerged as key contributors to ASD risk. Rare CNVs often demonstrate large effect sizes and can directly cause ASD, while common variants collectively exert subtle influences. Recent advances have identified numerous ASD-associated CNVs, including recurrent loci such as 1q21.1, 2p16.3, 7q11.23, 15q11.2, 15q11-q13, 16p11.2 and 22q11.2. Mouse models carrying these CNVs have provided profound insights into the underlying neurobiological mechanisms. Recent studies integrating transcriptomic, proteomic, and functional imaging approaches have revealed alterations in synaptic function, neuronal differentiation, myelination, metabolic pathways, and circuit connectivity. Notably, investigations leveraging conditional knockout models, high magnetic field MRI, and single-cell analyses highlight disruptions in excitatory-inhibitory balance, white matter integrity, and dynamic gene regulatory networks. Parallel human-based approaches, including iPSC-derived neurons, cerebral organoids, and large-scale single-nucleus sequencing, are combined with animal model data. These integrative strategies promise to refine our understanding of ASD's genetic architecture, bridging the gap between fundamental discoveries in model organisms and clinically relevant biomarkers, subtypes, and therapeutic targets in humans. This review summarizes key findings from recent CNV mouse model studies and highlights emerging technologies applied to human ASD samples. Finally, we outline prospects for translating findings from mouse studies to humans. By illuminating both unique and convergent genetic mechanisms, these advances offer a critical framework for unraveling etiological complexity in ASD.
Collapse
Affiliation(s)
- Kota Tamada
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, Kobe 650-0017, Japan.
| | - Toru Takumi
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, Kobe 650-0017, Japan.
| |
Collapse
|
3
|
Toolan KP, McGrath BT, Brinkmeier ML, Camper SA, Bielas SL. Ash1l loss-of-function results in structural birth defects and altered cortical development. Brain 2025; 148:55-68. [PMID: 38943682 PMCID: PMC11706301 DOI: 10.1093/brain/awae218] [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: 01/08/2024] [Revised: 05/16/2024] [Accepted: 06/09/2024] [Indexed: 07/01/2024] Open
Abstract
The histone methyltransferase ASH1L plays a crucial role in regulating gene expression across various organ systems during development, yet its role in brain development remains largely unexplored. Over 130 individuals with autism harbour heterozygous loss-of-function ASH1L variants, and population studies confirm it as a high-risk autism gene. Previous studies on Ash1l deficient mice have reported autistic-like behaviours and provided insights into the underlying neuropathophysiology. In this study, we used mice with a cre-inducible deletion of Ash1l exon 4, which results in a frame shift and premature stop codon (p.V1693Afs*2). Our investigation evaluated the impact of Ash1l loss-of-function on survival and craniofacial skeletal development. Using a tamoxifen-inducible cre strain, we targeted Ash1l knockout early in cortical development [Emx1-Cre-ERT2; embryonic Day (e) 10.5]. Immunohistochemistry was utilized to assess cortical lamination, while EdU incorporation aided in birthdating cortical neurons. Additionally, single-cell RNA sequencing was employed to compare cortical cell populations and identify genes with differential expression. At e18.5, the proportion of homozygous Ash1l germline knockout embryos appeared normal; however, no live Ash1l null pups were present at birth (e18.5: n = 77, P = 0.90; p0: n = 41, P = 0.00095). Notably, Ash1l-/- exhibited shortened nasal bones (n = 31, P = 0.017). In the cortical-specific knockout model, SATB2 neurons showed increased numbers (n = 6/genotype, P = 0.0001) and were distributed through the cortical plate. Birthdating revealed generation of ectopically placed deep layer neurons that express SATB2 (e13.5 injection: n = 4/genotype, P = 0.0126). Single cell RNA sequencing revealed significant differences in gene expression between control and mutant upper layer neurons, leading to distinct clustering. Pseudotime analysis indicated that the mutant cluster followed an altered cell differentiation trajectory. This study underscores the essential role of Ash1l in postnatal survival and normal craniofacial development. In the cortex, ASH1L exerts broad effects on gene expression and is indispensable for determining the fate of upper layer cortical neurons. These findings provide valuable insights into the potential mechanisms of ASH1L neuropathology, shedding light on its significance in neurodevelopmental disorders like autism.
Collapse
Affiliation(s)
- Kevin P Toolan
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI48109, USA
| | - Brian T McGrath
- Cell and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Michelle L Brinkmeier
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI48109, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI48109, USA
- Cell and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stephanie L Bielas
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI48109, USA
- Cell and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
4
|
Zhao Z, Okada N, Yagishita S, Yahata N, Nitta N, Shibata S, Abe Y, Morita S, Kumagai E, Tanaka KF, Suhara T, Takumi T, Kasai K, Jinde S. Correlations of brain structure with the social behavior of 15q11-13 duplication mice, an animal model of autism. Neurosci Res 2024; 209:42-49. [PMID: 39097003 DOI: 10.1016/j.neures.2024.07.009] [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: 02/21/2024] [Revised: 06/07/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Duplication of chromosome 15q11-13 has been reported to be one of the most frequent cytogenetic copy number variations in autism spectrum disorder (ASD), and a mouse model of paternal 15q11-13 duplication was generated, termed 15q dup mice. While previous studies have replicated some of the behavioral and brain structural phenotypes of ASD separately, the relationship between brain structure and behavior has rarely been examined. In this study, we performed behavioral experiments related to anxiety and social behaviors and magnetic resonance imaging (MRI) using the same set of 15q dup and wild-type mice. 15q dup mice showed increased anxiety and a tendency toward alterations in social behaviors, as reported previously, as well as variability in terms of sociability. MRI analysis revealed that a lower sociability index was correlated with a smaller gray matter volume in the right medial entorhinal cortex. These results may help to understand how variability in behavioral phenotypes of ASD arises even in individuals with the same genetic background and to determine the individual differences in neurodevelopmental trajectory correlated with specific brain structures that underlie these phenotypes.
Collapse
Affiliation(s)
- Zhilei Zhao
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan; International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naohiro Okada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan; International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Sho Yagishita
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan; Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, Faculty of Medicine Bldg, The University of Tokyo, 1 #NC207, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Noriaki Yahata
- National Institutes for Quantum Sciences and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Nobuhiro Nitta
- National Institutes for Quantum Sciences and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan; Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki Ward, Kawasaki, Kanagawa 210-0821, Japan
| | - Sayaka Shibata
- National Institutes for Quantum Sciences and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Yoshifumi Abe
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Susumu Morita
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Eureka Kumagai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Tetsuya Suhara
- National Institutes for Quantum Sciences and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Toru Takumi
- Department of Physiology and Cell Biology, Kobe University School of Medicine, 7-5-1 Kusunoki-cho, Chuo, Kobe 650-0017, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan; International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Seiichiro Jinde
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan.
| |
Collapse
|
5
|
Forrest MP, Penzes P. Mechanisms of copy number variants in neuropsychiatric disorders: From genes to therapeutics. Curr Opin Neurobiol 2023; 82:102750. [PMID: 37515924 PMCID: PMC10529795 DOI: 10.1016/j.conb.2023.102750] [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: 03/12/2023] [Revised: 06/01/2023] [Accepted: 06/27/2023] [Indexed: 07/31/2023]
Abstract
Copy number variants (CNVs) are genomic imbalances strongly linked to the aetiology of neuropsychiatric disorders such as schizophrenia and autism. By virtue of their large size, CNVs often contain many genes, providing a multi-genic view of disease processes that can be dissected in model systems. Thus, CNV research provides an important stepping stone towards understanding polygenic disease mechanisms, positioned between monogenic and polygenic risk models. In this review, we will outline hypothetical models for gene interactions occurring within CNVs and discuss different approaches used to study rodent and stem cell disease models. We highlight recent work showing that genetic and pharmacological strategies can be used to rescue important aspects of CNV-mediated pathophysiology, which often converges onto synaptic pathways. We propose that using a rescue approach in complete CNV models provides a new path forward for precise mechanistic understanding of complex disorders and a tangible route towards therapeutic development.
Collapse
Affiliation(s)
- Marc P Forrest
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Peter Penzes
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
6
|
Zhang J, Cai F, Lu R, Xing X, Xu L, Wu K, Gong Z, Zhang Q, Zhang Y, Xing M, Song W, Li JD. CNTNAP2 intracellular domain (CICD) generated by γ-secretase cleavage improves autism-related behaviors. Signal Transduct Target Ther 2023; 8:219. [PMID: 37271769 DOI: 10.1038/s41392-023-01431-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/03/2023] [Accepted: 03/24/2023] [Indexed: 06/06/2023] Open
Abstract
As the most prevalent neurodevelopmental disorders in children, autism spectrum disorders (ASD) are characterized by deficits in language development, social interaction, and repetitive behaviors or inflexible interests. Contactin associated protein like 2 (CNTNAP2), encoding a single transmembrane protein (CNTNAP2) with 1331 amino acid residues, is a widely validated ASD-susceptible gene. Cntnap2-deficient mice also show core autism-relevant behaviors, including the social deficits and repetitive behavior. However, the cellular mechanisms underlying dysfunction CNTNAP2 and ASD remain elusive. In this study, we found a motif within the transmembrane domain of CNTNAP2 was highly homologous to the γ-secretase cleavage site of amyloid-β precursor protein (APP), suggesting that CNTNAP2 may undergo proteolytic cleavage. Further biochemical analysis indicated that CNTNAP2 is cleaved by γ-secretase to produce the CNTNAP2 intracellular domain (CICD). Virally delivery of CICD to the medial prefrontal cortex (mPFC) in Cntnap2-deficient (Cntnap2-/-) mice normalized the deficit in the ASD-related behaviors, including social deficit and repetitive behaviors. Furthermore, CICD promoted the nuclear translocation of calcium/calmodulin-dependent serine protein kinase (CASK) to regulate the transcription of genes, such as Prader Willi syndrome gene Necdin. Whereas Necdin deficiency led to reduced social interaction in mice, virally expression of Necdin in the mPFC normalized the deficit in social preference of Cntnap2-/- mice. Our results thus reveal a critical function of CICD and highlight a role of the CNTNAP2-CASK-Necdin signaling pathway in ASD.
Collapse
Affiliation(s)
- Jing Zhang
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Fang Cai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Renbin Lu
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Xiaoliang Xing
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Lu Xu
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Kunyang Wu
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Zishan Gong
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Qing Zhang
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yun Zhang
- Advanced Innovation Center for Human Brain Protection, The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Mengen Xing
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Weihong Song
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Jia-Da Li
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
7
|
Nakai N, Sato M, Yamashita O, Sekine Y, Fu X, Nakai J, Zalesky A, Takumi T. Virtual reality-based real-time imaging reveals abnormal cortical dynamics during behavioral transitions in a mouse model of autism. Cell Rep 2023; 42:112258. [PMID: 36990094 DOI: 10.1016/j.celrep.2023.112258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/16/2023] [Accepted: 02/28/2023] [Indexed: 03/30/2023] Open
Abstract
Functional connectivity (FC) can provide insight into cortical circuit dysfunction in neuropsychiatric disorders. However, dynamic changes in FC related to locomotion with sensory feedback remain to be elucidated. To investigate FC dynamics in locomoting mice, we develop mesoscopic Ca2+ imaging with a virtual reality (VR) environment. We find rapid reorganization of cortical FC in response to changing behavioral states. By using machine learning classification, behavioral states are accurately decoded. We then use our VR-based imaging system to study cortical FC in a mouse model of autism and find that locomotion states are associated with altered FC dynamics. Furthermore, we identify FC patterns involving the motor area as the most distinguishing features of the autism mice from wild-type mice during behavioral transitions, which might correlate with motor clumsiness in individuals with autism. Our VR-based real-time imaging system provides crucial information to understand FC dynamics linked to behavioral abnormality of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Nobuhiro Nakai
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan; Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, Kobe 650-0017, Japan
| | - Masaaki Sato
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan; Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Kita, Sapporo 060-8638, Japan.
| | - Okito Yamashita
- RIKEN Center for Advanced Intelligence Project, Chuo, Tokyo 103-0027, Japan; Department of Computational Brain Imaging, ATR Neural Information Analysis Laboratories, Seika, Kyoto 619-0288, Japan
| | - Yukiko Sekine
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Xiaochen Fu
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Junichi Nakai
- Division of Oral Physiology, Department of Disease Management Dentistry, Tohoku University Graduate School of Dentistry, Aoba, Sendai 980-8575, Japan
| | - Andrew Zalesky
- Melbourne Neuropsychiatry Centre and Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Toru Takumi
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan; Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, Kobe 650-0017, Japan; RIKEN Center for Biosystems Dynamics Research, Chuo, Kobe 650-0047, Japan.
| |
Collapse
|
8
|
Elamin M, Dumarchey A, Stoddard C, Robinson TM, Cowie C, Gorka D, Chamberlain SJ, Levine ES. The role of UBE3A in the autism and epilepsy-related Dup15q syndrome using patient-derived, CRISPR-corrected neurons. Stem Cell Reports 2023; 18:884-898. [PMID: 36898382 PMCID: PMC10147551 DOI: 10.1016/j.stemcr.2023.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 03/11/2023] Open
Abstract
Chromosome 15q11-q13 duplication syndrome (Dup15q) is a neurodevelopmental disorder caused by maternal duplications of this region. Autism and epilepsy are key features of Dup15q. UBE3A, which encodes an E3 ubiquitin ligase, is likely a major driver of Dup15q because UBE3A is the only imprinted gene expressed solely from the maternal allele. Nevertheless, the exact role of UBE3A has not been determined. To establish whether UBE3A overexpression is required for Dup15q neuronal deficits, we generated an isogenic control line for a Dup15q patient-derived induced pluripotent stem cell line. Dup15q neurons exhibited hyperexcitability compared with control neurons, and this phenotype was generally prevented by normalizing UBE3A levels using antisense oligonucleotides. Overexpression of UBE3A resulted in a profile similar to that of Dup15q neurons except for synaptic phenotypes. These results indicate that UBE3A overexpression is necessary for most Dup15q cellular phenotypes but also suggest a role for other genes in the duplicated region.
Collapse
Affiliation(s)
- Marwa Elamin
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Aurelie Dumarchey
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Christopher Stoddard
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Tiwanna M Robinson
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Christopher Cowie
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Dea Gorka
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Stormy J Chamberlain
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA.
| |
Collapse
|
9
|
Punt AM, Judson MC, Sidorov MS, Williams BN, Johnson NS, Belder S, den Hertog D, Davis CR, Feygin MS, Lang PF, Jolfaei MA, Curran PJ, van IJcken WF, Elgersma Y, Philpot BD. Molecular and behavioral consequences of Ube3a gene overdosage in mice. JCI Insight 2022; 7:e158953. [PMID: 36134658 PMCID: PMC9675564 DOI: 10.1172/jci.insight.158953] [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: 01/31/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022] Open
Abstract
Chromosome 15q11.2-q13.1 duplication syndrome (Dup15q syndrome) is a severe neurodevelopmental disorder characterized by intellectual disability, impaired motor coordination, and autism spectrum disorder. Chromosomal multiplication of the UBE3A gene is presumed to be the primary driver of Dup15q pathophysiology, given that UBE3A exhibits maternal monoallelic expression in neurons and that maternal duplications typically yield far more severe neurodevelopmental outcomes than paternal duplications. However, studies into the pathogenic effects of UBE3A overexpression in mice have yielded conflicting results. Here, we investigated the neurodevelopmental impact of Ube3a gene overdosage using bacterial artificial chromosome-based transgenic mouse models (Ube3aOE) that recapitulate the increases in Ube3a copy number most often observed in Dup15q. In contrast to previously published Ube3a overexpression models, Ube3aOE mice were indistinguishable from wild-type controls on a number of molecular and behavioral measures, despite suffering increased mortality when challenged with seizures, a phenotype reminiscent of sudden unexpected death in epilepsy. Collectively, our data support a model wherein pathogenic synergy between UBE3A and other overexpressed 15q11.2-q13.1 genes is required for full penetrance of Dup15q syndrome phenotypes.
Collapse
Affiliation(s)
- A. Mattijs Punt
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Matthew C. Judson
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Michael S. Sidorov
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Brittany N. Williams
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Naomi S. Johnson
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Sabine Belder
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Dion den Hertog
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Courtney R. Davis
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Maximillian S. Feygin
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Patrick F. Lang
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Mehrnoush Aghadavoud Jolfaei
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Patrick J. Curran
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Ype Elgersma
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Benjamin D. Philpot
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| |
Collapse
|
10
|
Zhou F, Ebea P, Mutai E, Wang H, Sukreet S, Navazesh S, Dogan H, Li W, Cui J, Ji P, Ramirez DMO, Zempleni J. Small Extracellular Vesicles in Milk Cross the Blood-Brain Barrier in Murine Cerebral Cortex Endothelial Cells and Promote Dendritic Complexity in the Hippocampus and Brain Function in C57BL/6J Mice. Front Nutr 2022; 9:838543. [PMID: 35600828 PMCID: PMC9121399 DOI: 10.3389/fnut.2022.838543] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/29/2022] [Indexed: 12/12/2022] Open
Abstract
Human milk contains large amounts of small extracellular vesicles (sEVs) and their microRNA cargos, whereas infant formulas contain only trace amounts of sEVs and microRNAs. We assessed the transport of sEVs across the blood-brain barrier (BBB) and sEV accumulation in distinct regions of the brain in brain endothelial cells and suckling mice. We further assessed sEV-dependent gene expression profiles and effects on the dendritic complexity of hippocampal granule cells and phenotypes of EV depletion in neonate, juvenile and adult mice. The transfer of sEVs across the BBB was assessed by using fluorophore-labeled bovine sEVs in brain endothelial bEnd.3 monolayers and dual chamber systems, and in wild-type newborn pups fostered to sEV and cargo tracking (ECT) dams that express sEVs labeled with a CD63-eGFP fusion protein for subsequent analysis by serial two-photon tomography and staining with anti-eGFP antibodies. Effects of EVs on gene expression and dendritic architecture of granule cells was analyzed in hippocampi from juvenile mice fed sEV and RNA-depleted (ERD) and sEV and RNA-sufficient (ERS) diets by using RNA-sequencing analysis and Golgi-Cox staining followed by integrated neuronal tracing and morphological analysis of neuronal dendrites, respectively. Spatial learning and severity of kainic acid-induced seizures were assessed in mice fed ERD and ERS diets. bEnd.3 cells internalized sEVs by using a saturable transport mechanism and secreted miR-34a across the basal membrane. sEVs penetrated the entire brain in fostering experiments; major regions of accumulation included the hippocampus, cortex and cerebellum. Two hundred ninety-five genes were differentially expressed in hippocampi from mice fed ERD and ERS diets; high-confidence gene networks included pathways implicated in axon guidance and calcium signaling. Juvenile pups fed the ERD diet had reduced dendritic complexity of dentate granule cells in the hippocampus, scored nine-fold lower in the Barnes maze test of spatial learning and memory, and the severity of seizures was 5-fold higher following kainic acid administration in adult mice fed the ERD diet compared to mice fed the ERS diet. We conclude that sEVs cross the BBB and contribute toward optimal neuronal development, spatial learning and memory, and resistance to kainic acid-induced seizures in mice.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Pearl Ebea
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Ezra Mutai
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Haichuan Wang
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Sonal Sukreet
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Shya Navazesh
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Haluk Dogan
- School of Computing, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Wenhao Li
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Juan Cui
- School of Computing, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Peng Ji
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Denise M. O. Ramirez
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|