51
|
Abstract
Recent advances in genomics have revealed a wide spectrum of genetic variants associated with neurodevelopmental disorders at an unprecedented scale. An increasing number of studies have consistently identified mutations-both inherited and de novo-impacting the function of specific brain circuits. This suggests that, during brain development, alterations in distinct neural circuits, cell types, or broad regulatory pathways ultimately shaping synapses might be a dysfunctional process underlying these disorders. Here, we review findings from human studies and animal model research to provide a comprehensive description of synaptic and circuit mechanisms implicated in neurodevelopmental disorders. We discuss how specific synaptic connections might be commonly disrupted in different disorders and the alterations in cognition and behaviors emerging from imbalances in neuronal circuits. Moreover, we review new approaches that have been shown to restore or mitigate dysfunctional processes during specific critical windows of brain development. Considering the heterogeneity of neurodevelopmental disorders, we also highlight the recent progress in developing improved clinical biomarkers and strategies that will help to identify novel therapeutic compounds and opportunities for early intervention.
Collapse
Affiliation(s)
- David Exposito-Alonso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom;
- Current affiliation: Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| | - Beatriz Rico
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom;
| |
Collapse
|
52
|
Dong C, Zhao C, Chen X, Berry K, Wang J, Zhang F, Liao Y, Han R, Ogurek S, Xu L, Zhang L, Lin Y, Zhou W, Xin M, Lim DA, Campbell K, Nakafuku M, Waclaw RR, Lu QR. Conserved and Distinct Functions of the Autism-Related Chromatin Remodeler CHD8 in Embryonic and Adult Forebrain Neurogenesis. J Neurosci 2022; 42:8373-8392. [PMID: 36127134 PMCID: PMC9653284 DOI: 10.1523/jneurosci.2400-21.2022] [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: 12/04/2021] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 11/21/2022] Open
Abstract
The chromatin remodeler CHD8 represents a high-confidence risk factor in autism, a multistage progressive neurologic disorder, however the underlying stage-specific functions remain elusive. In this study, by analyzing Chd8 conditional knock-out mice (male and female), we find that CHD8 controls cortical neural stem/progenitor cell (NSC) proliferation and survival in a stage-dependent manner. Strikingly, inducible genetic deletion reveals that CHD8 is required for the production and fitness of transit-amplifying intermediate progenitors (IPCs) essential for upper-layer neuron expansion in the embryonic cortex. p53 loss of function partially rescues apoptosis and neurogenesis defects in the Chd8-deficient brain. Further, transcriptomic and epigenomic profiling indicates that CHD8 regulates the chromatin accessibility landscape to activate neurogenesis-promoting factors including TBR2, a key regulator of IPC neurogenesis, while repressing DNA damage- and p53-induced apoptotic programs. In the adult brain, CHD8 depletion impairs forebrain neurogenesis by impeding IPC differentiation from NSCs in both subventricular and subgranular zones; however, unlike in embryos, it does not affect NSC proliferation and survival. Treatment with an antidepressant approved by the Federal Drug Administration (FDA), fluoxetine, partially restores adult hippocampal neurogenesis in Chd8-ablated mice. Together, our multistage functional studies identify temporally specific roles for CHD8 in developmental and adult neurogenesis, pointing to a potential strategy to enhance neurogenesis in the CHD8-deficient brain.SIGNIFICANCE STATEMENT The role of the high-confidence autism gene CHD8 in neurogenesis remains incompletely understood. Here, we identify a stage-specific function of CHD8 in development of NSCs in developing and adult brains by conserved, yet spatiotemporally distinct, mechanisms. In embryonic cortex, CHD8 is critical for the proliferation, survival, and differentiation of both NSC and IPCs during cortical neurogenesis. In adult brain, CHD8 is required for IPC generation but not the proliferation and survival of adult NSCs. Treatment with FDA-approved antidepressant fluoxetine partially rescues the adult neurogenesis defects in CHD8 mutants. Thus, our findings help resolve CHD8 functions throughout life during embryonic and adult neurogenesis and point to a potential avenue to promote neurogenesis in CHD8 deficiency.
Collapse
Affiliation(s)
- Chen Dong
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Chuntao Zhao
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Xiang Chen
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Kalen Berry
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Jiajia Wang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Feng Zhang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Yunfei Liao
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Rong Han
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Sean Ogurek
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Lingli Xu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Li Zhang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Yifeng Lin
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wenhao Zhou
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Mei Xin
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Daniel A Lim
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California 94143
| | - Kenneth Campbell
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Masato Nakafuku
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Ronald R Waclaw
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| |
Collapse
|
53
|
Yang D, Zhao Y, Nie B, An L, Wan X, Wang Y, Wang W, Cai G, Wu S. Progress in magnetic resonance imaging of autism model mice brain. WILEY INTERDISCIPLINARY REVIEWS. COGNITIVE SCIENCE 2022; 13:e1616. [PMID: 35930672 DOI: 10.1002/wcs.1616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/11/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disease characterized by social disorder and stereotypical behaviors with an increasing incidence. ASD patients are suffering from varying degrees of mental retardation and language development abnormalities. Magnetic resonance imaging (MRI) is a noninvasive imaging technology to detect brain structural and functional dysfunction in vivo, playing an important role in the early diagnosisbasic research of ASD. High-field, small-animal MRI in basic research of autism model mice has provided a new approach to research the pathogenesis, characteristics, and intervention efficacy in autism. This article reviews MRI studies of mouse models of autism over the past 20 years. Reduced gray matter, abnormal connections of brain networks, and abnormal development of white matter fibers have been demonstrated in these studies, which are present in different proportions in the various mouse models. This provides a more macroscopic view for subsequent research on autism model mice. This article is categorized under: Cognitive Biology > Genes and Environment Neuroscience > Computation Neuroscience > Genes, Molecules, and Cells Neuroscience > Development.
Collapse
Affiliation(s)
- Dingding Yang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yan Zhao
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Binbin Nie
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Leiting An
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xiangdong Wan
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yazhou Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Guohong Cai
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
54
|
Lee SY, Kweon H, Kang H, Kim E. Age-differential sexual dimorphism in CHD8-S62X-mutant mouse behaviors. Front Mol Neurosci 2022; 15:1022306. [DOI: 10.3389/fnmol.2022.1022306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorders (ASD) are ~4-times more common in males than females, and CHD8 (a chromatin remodeler)-related ASD shows a strong male bias (~4:1), although the underlying mechanism remains unclear. Chd8-mutant mice with a C-terminal protein-truncating mutation (N2373K) display male-preponderant behavioral deficits as juveniles and adults, although whether this also applies to other Chd8 mutations remains unknown. In addition, it remains unclear whether sexually dimorphic phenotypes in Chd8-mutant mice are differentially observed in males and females across different ages. We here generated new Chd8-mutant (knock-in) mice carrying a patient-derived mutation causing an N-terminal and stronger protein truncation (Chd8+/S62X mice) and characterized the mice by behavioral analyses. Juvenile Chd8+/S62X mice displayed male-preponderant autistic-like behaviors; hypoactivity and enhanced mother-seeking/attachment behavior in males but not in females. Adult male and female Chd8+/S62X mice showed largely similar deficits in repetitive and anxiety-like behavioral domains. Therefore, the CHD8-S62X mutation induces ASD-like behaviors in juvenile male mice and adult male and female mice, pointing to an age-differential sexual dimorphism and also distinct sexual dimorphisms in different Chd8 mutations (N2373K and S62X).
Collapse
|
55
|
Coakley-Youngs E, Ranatunga M, Richardson S, Getti G, Shorter S, Fivaz M. Autism-associated CHD8 keeps proliferation of human neural progenitors in check by lengthening the G1 phase of the cell cycle. Biol Open 2022; 11:276883. [PMID: 36222238 PMCID: PMC9548376 DOI: 10.1242/bio.058941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/28/2022] [Indexed: 01/17/2023] Open
Abstract
ABSTRACT
De novo mutations (DNMs) in chromodomain helicase DNA binding protein 8 (CHD8) are associated with a specific subtype of autism characterized by enlarged heads and distinct cranial features. The vast majority of these DNMs are heterozygous loss-of-function mutations with high penetrance for autism. CHD8 is a chromatin remodeler that preferentially regulates expression of genes implicated in early development of the cerebral cortex. How CHD8 haploinsufficiency alters the normal developmental trajectory of the brain is poorly understood and debated. Using long-term single-cell imaging, we show that disruption of a single copy of CHD8 in human neural precursor cells (NPCs) markedly shortens the G1 phase of the cell cycle. Consistent with faster progression of CHD8+/− NPCs through G1 and the G1/S checkpoint, we observed increased expression of E cyclins and elevated phosphorylation of Erk in these mutant cells – two central signaling pathways involved in S phase entry. Thus, CHD8 keeps proliferation of NPCs in check by lengthening G1, and mono-allelic disruption of this gene alters cell-cycle timing in a way that favors self-renewing over neurogenic cell divisions. Our findings further predict enlargement of the neural progenitor pool in CHD8+/− developing brains, providing a mechanistic basis for macrocephaly in this autism subtype.
Collapse
Affiliation(s)
- Emma Coakley-Youngs
- Stem Cell & Gene Editing Laboratory, University of Greenwich at Medway 1 , Faculty of Science and Engineering, Kent ME4 4TB , UK
| | - Medhavi Ranatunga
- University of Greenwich at Medway 2 , Faculty of Science and Engineering, Kent ME4 4TB , UK
| | - Simon Richardson
- Exogenics Laboratory, University of Greenwich at Medway 3 , Faculty of Science and Engineering, Kent ME4 4TB , UK
| | - Giulia Getti
- University of Greenwich at Medway 2 , Faculty of Science and Engineering, Kent ME4 4TB , UK
| | - Susan Shorter
- Stem Cell & Gene Editing Laboratory, University of Greenwich at Medway 1 , Faculty of Science and Engineering, Kent ME4 4TB , UK
| | - Marc Fivaz
- Stem Cell & Gene Editing Laboratory, University of Greenwich at Medway 1 , Faculty of Science and Engineering, Kent ME4 4TB , UK
| |
Collapse
|
56
|
Npas3 deficiency impairs cortical astrogenesis and induces autistic-like behaviors. Cell Rep 2022; 40:111289. [PMID: 36044858 DOI: 10.1016/j.celrep.2022.111289] [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: 12/08/2021] [Revised: 06/13/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022] Open
Abstract
Transcription factors with basic-helix-loop-helix (bHLH) motifs can control neural progenitor fate determination to neurons and oligodendrocytes. How bHLH transcription factors regulate astrogenesis remains largely unknown. Here, we report that NPAS3, a bHLH transcription factor, is a critical regulator of astrogenesis. Npas3 deficiency impairs cortical astrogenesis, correlating with abnormal brain development and autistic-like behaviors. Single-cell transcriptomes reveal that Npas3 knockout induces abnormal transition states in the differentiation trajectories from radial glia to astrocytes. Analysis of chromatin immunoprecipitation sequencing data in primary cortical astrocytes shows that NPAS3 binding targets are involved in functions of brain development and synapse organization. Co-culture assay further indicates that NPAS3-impaired astrogenesis induces synaptic deficits in wild-type neurons. Astrocyte-specific knockdown of NPAS3 in wild-type cortex causes synaptic and behavioral abnormalities associated with the core symptoms in autism. Together, our findings suggest that transcription factor NPAS3 regulates astrogenesis and its subsequent consequences for brain development and behavior.
Collapse
|
57
|
Al-Amri AH, Armstrong P, Amici M, Ligneul C, Rouse J, El-Asrag ME, Pantiru A, Vancollie VE, Ng HW, Ogbeta JA, Goodchild K, Ellegood J, Lelliott CJ, Mullins JG, Bretman A, Al-Ali R, Beetz C, Al-Gazali L, Al Shamsi A, Lerch JP, Mellor JR, Al Sayegh A, Ali M, Inglehearn CF, Clapcote SJ. PDZD8 Disruption Causes Cognitive Impairment in Humans, Mice, and Fruit Flies. Biol Psychiatry 2022; 92:323-334. [PMID: 35227461 PMCID: PMC9302898 DOI: 10.1016/j.biopsych.2021.12.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND The discovery of coding variants in genes that confer risk of intellectual disability (ID) is an important step toward understanding the pathophysiology of this common developmental disability. METHODS Homozygosity mapping, whole-exome sequencing, and cosegregation analyses were used to identify gene variants responsible for syndromic ID with autistic features in two independent consanguineous families from the Arabian Peninsula. For in vivo functional studies of the implicated gene's function in cognition, Drosophila melanogaster and mice with targeted interference of the orthologous gene were used. Behavioral, electrophysiological, and structural magnetic resonance imaging analyses were conducted for phenotypic testing. RESULTS Homozygous premature termination codons in PDZD8, encoding an endoplasmic reticulum-anchored lipid transfer protein, showed cosegregation with syndromic ID in both families. Drosophila melanogaster with knockdown of the PDZD8 ortholog exhibited impaired long-term courtship-based memory. Mice homozygous for a premature termination codon in Pdzd8 exhibited brain structural, hippocampal spatial memory, and synaptic plasticity deficits. CONCLUSIONS These data demonstrate the involvement of homozygous loss-of-function mutations in PDZD8 in a neurodevelopmental cognitive disorder. Model organisms with manipulation of the orthologous gene replicate aspects of the human phenotype and suggest plausible pathophysiological mechanisms centered on disrupted brain development and synaptic function. These findings are thus consistent with accruing evidence that synaptic defects are a common denominator of ID and other neurodevelopmental conditions.
Collapse
Affiliation(s)
- Ahmed H. Al-Amri
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom,Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom,National Genetic Centre, Royal Hospital, Muscat, Oman
| | - Paul Armstrong
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Mascia Amici
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Clemence Ligneul
- Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, United Kingdom
| | - James Rouse
- School of Biology, University of Leeds, Leeds, United Kingdom
| | - Mohammed E. El-Asrag
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom,Department of Zoology, Faculty of Science, Benha University, Benha, Egypt,Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham
| | - Andreea Pantiru
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Hannah W.Y. Ng
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Jennifer A. Ogbeta
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Kirstie Goodchild
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | - Amanda Bretman
- School of Biology, University of Leeds, Leeds, United Kingdom
| | | | | | - Lihadh Al-Gazali
- Department of Paediatrics, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Aisha Al Shamsi
- Pediatrics Department, Tawam Hospital, Al Ain, United Arab Emirates
| | - Jason P. Lerch
- Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, United Kingdom
| | - Jack R. Mellor
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Abeer Al Sayegh
- Genetics Department, Sultan Qaboos University Hospital, Muscat, Oman
| | - Manir Ali
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Chris F. Inglehearn
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Steven J. Clapcote
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom,Address correspondence to Steven J. Clapcote, Ph.D.
| |
Collapse
|
58
|
Dougnon G, Matsui H. Modelling Autism Spectrum Disorder (ASD) and Attention-Deficit/Hyperactivity Disorder (ADHD) Using Mice and Zebrafish. Int J Mol Sci 2022; 23:ijms23147550. [PMID: 35886894 PMCID: PMC9319972 DOI: 10.3390/ijms23147550] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorders (ASD) and attention-deficit/hyperactivity disorder (ADHD) are two debilitating neurodevelopmental disorders. The former is associated with social impairments whereas the latter is associated with inattentiveness, hyperactivity, and impulsivity. There is recent evidence that both disorders are somehow related and that genes may play a large role in these disorders. Despite mounting human and animal research, the neurological pathways underlying ASD and ADHD are still not well understood. Scientists investigate neurodevelopmental disorders by using animal models that have high similarities in genetics and behaviours with humans. Mice have been utilized in neuroscience research as an excellent animal model for a long time; however, the zebrafish has attracted much attention recently, with an increasingly large number of studies using this model. In this review, we first discuss ASD and ADHD aetiology from a general point of view to their characteristics and treatments. We also compare mice and zebrafish for their similarities and discuss their advantages and limitations in neuroscience. Finally, we summarize the most recent and existing research on zebrafish and mouse models of ASD and ADHD. We believe that this review will serve as a unique document providing interesting information to date about these models, thus facilitating research on ASD and ADHD.
Collapse
|
59
|
Adhikari A, Buchanan FKB, Fenton TA, Cameron DL, Halmai JANM, Copping NA, Fink KD, Silverman JL. Touchscreen Cognitive Deficits, Hyperexcitability, and Hyperactivity in Males and Females Using Two Models of Cdkl5 Deficiency. Hum Mol Genet 2022; 31:3032-3050. [PMID: 35445702 PMCID: PMC9476626 DOI: 10.1093/hmg/ddac091] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/17/2022] Open
Abstract
Many neurodevelopmental disorders (NDDs) are the result of mutations on the X chromosome. One severe NDD resulting from mutations on the X chromosome is CDKL5 deficiency disorder (CDD). CDD is an epigenetic, X-linked NDD characterized by intellectual disability (ID), pervasive seizures and severe sleep disruption, including recurring hospitalizations. CDD occurs at a 4:1 ratio, with a female bias. CDD is driven by the loss of cyclin-dependent kinase-like 5 (CDKL5), a serine/threonine kinase that is essential for typical brain development, synapse formation and signal transmission. Previous studies focused on male subjects from animal models, likely to avoid the complexity of X mosaicism. For the first time, we report translationally relevant behavioral phenotypes in young adult (8–20 weeks) females and males with robust signal size, including impairments in learning and memory, substantial hyperactivity and increased susceptibility to seizures/reduced seizure thresholds, in both sexes, and in two models of CDD preclinical mice, one with a general loss-of-function mutation and one that is a patient-derived mutation.
Collapse
Affiliation(s)
- Anna Adhikari
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA
| | - Fiona K B Buchanan
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA.,Stem Cell Program and Gene Therapy Center, University of California Davis School of Medicine, Sacramento, CA
| | - Timothy A Fenton
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA
| | - David L Cameron
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA.,Stem Cell Program and Gene Therapy Center, University of California Davis School of Medicine, Sacramento, CA
| | - Julian A N M Halmai
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA.,Stem Cell Program and Gene Therapy Center, University of California Davis School of Medicine, Sacramento, CA
| | - Nycole A Copping
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA
| | - Kyle D Fink
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA.,Department of Neurology, University of California Davis School of Medicine, Sacramento, CA.,Stem Cell Program and Gene Therapy Center, University of California Davis School of Medicine, Sacramento, CA
| | - Jill L Silverman
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA
| |
Collapse
|
60
|
Villa CE, Cheroni C, Dotter CP, López-Tóbon A, Oliveira B, Sacco R, Yahya AÇ, Morandell J, Gabriele M, Tavakoli MR, Lyudchik J, Sommer C, Gabitto M, Danzl JG, Testa G, Novarino G. CHD8 haploinsufficiency links autism to transient alterations in excitatory and inhibitory trajectories. Cell Rep 2022; 39:110615. [PMID: 35385734 DOI: 10.1016/j.celrep.2022.110615] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 11/18/2021] [Accepted: 03/13/2022] [Indexed: 12/13/2022] Open
Abstract
Mutations in the chromodomain helicase DNA-binding 8 (CHD8) gene are a frequent cause of autism spectrum disorder (ASD). While its phenotypic spectrum often encompasses macrocephaly, implicating cortical abnormalities, how CHD8 haploinsufficiency affects neurodevelopmental is unclear. Here, employing human cerebral organoids, we find that CHD8 haploinsufficiency disrupted neurodevelopmental trajectories with an accelerated and delayed generation of, respectively, inhibitory and excitatory neurons that yields, at days 60 and 120, symmetrically opposite expansions in their proportions. This imbalance is consistent with an enlargement of cerebral organoids as an in vitro correlate of patients' macrocephaly. Through an isogenic design of patient-specific mutations and mosaic organoids, we define genotype-phenotype relationships and uncover their cell-autonomous nature. Our results define cell-type-specific CHD8-dependent molecular defects related to an abnormal program of proliferation and alternative splicing. By identifying cell-type-specific effects of CHD8 mutations, our study uncovers reproducible developmental alterations that may be employed for neurodevelopmental disease modeling.
Collapse
Affiliation(s)
- Carlo Emanuele Villa
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy
| | - Cristina Cheroni
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| | - Christoph P Dotter
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Alejandro López-Tóbon
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| | - Bárbara Oliveira
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Roberto Sacco
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Aysan Çerağ Yahya
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Jasmin Morandell
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Michele Gabriele
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy
| | - Mojtaba R Tavakoli
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Julia Lyudchik
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Christoph Sommer
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | | | - Johann G Danzl
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Giuseppe Testa
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy.
| | - Gaia Novarino
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria.
| |
Collapse
|
61
|
Jiménez JA, Simon JM, Hu W, Moy SS, Harper KM, Liu CW, Lu K, Zylka MJ. Developmental pyrethroid exposure and age influence phenotypes in a Chd8 haploinsufficient autism mouse model. Sci Rep 2022; 12:5555. [PMID: 35365720 PMCID: PMC8975859 DOI: 10.1038/s41598-022-09533-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/24/2022] [Indexed: 11/08/2022] Open
Abstract
Hundreds of genes have been associated with autism spectrum disorder (ASD), including loss-of-function mutations in chromodomain helicase DNA binding protein 8 (Chd8). Environmental factors also are implicated in autism risk and have the potential to exacerbate phenotypes in genetically sensitized backgrounds. Here we investigate transcriptional and behavioral phenotypes in a Chd8 haploinsufficient (Chd8V986*/+) mouse line exposed to the pesticide deltamethrin (DM) from conception to postnatal day 22. Vehicle-exposed Chd8V986*/+ mice displayed ASD-associated phenotypes, including anxiety-like behavior and altered sociability, replicating a previous study with this mouse line. A core set of genes was altered in Chd8V986*/+ mice at multiple ages, including Usp11, Wars2, Crlf2, and Eglf6, and proximity ligation data indicated direct binding of CHD8 to the 5' region of these genes. Moreover, oligodendrocyte and neurodegenerative transcriptional phenotypes were apparent in 12 and 18 month old Chd8V986*/+ mice. Following DM exposure, the mutant mice displayed an exacerbated phenotype in the elevated plus maze, and genes associated with vascular endothelial cells were downregulated in the cerebral cortex of older Chd8V986*/+ animals. Our study reveals a gene x environment interaction with a Chd8 haploinsufficient mouse line and points to the importance of investigating phenotypes in ASD animal models across the lifespan.
Collapse
Affiliation(s)
- Jessica A Jiménez
- Curriculum in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jeremy M Simon
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Wenxin Hu
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sheryl S Moy
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kathryn M Harper
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chih-Wei Liu
- Department of Environmental Sciences and Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kun Lu
- Department of Environmental Sciences and Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mark J Zylka
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
62
|
Silverman JL, Thurm A, Ethridge SB, Soller MM, Petkova SP, Abel T, Bauman MD, Brodkin ES, Harony‐Nicolas H, Wöhr M, Halladay A. Reconsidering animal models used to study autism spectrum disorder: Current state and optimizing future. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12803. [PMID: 35285132 PMCID: PMC9189007 DOI: 10.1111/gbb.12803] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 12/15/2022]
Abstract
Neurodevelopmental disorders (NDDs), including autism spectrum disorder (ASD) and intellectual disability (ID), are pervasive, often lifelong disorders, lacking evidence-based interventions for core symptoms. With no established biological markers, diagnoses are defined by behavioral criteria. Thus, preclinical in vivo animal models of NDDs must be optimally utilized. For this reason, experts in the field of behavioral neuroscience convened a workshop with the goals of reviewing current behavioral studies, reports, and assessments in rodent models. Goals included: (a) identifying the maximal utility and limitations of behavior in animal models with construct validity; (b) providing recommendations for phenotyping animal models; and (c) guidelines on how in vivo models should be used and reported reliably and rigorously while acknowledging their limitations. We concluded by recommending minimal criteria for reporting in manuscripts going forward. The workshop elucidated a consensus of potential solutions to several problems, including revisiting claims made about animal model links to ASD (and related conditions). Specific conclusions included: mice (or other rodent or preclinical models) are models of the neurodevelopmental insult, not specifically any disorder (e.g., ASD); a model that perfectly recapitulates a disorder such as ASD is untenable; and greater attention needs be given to validation of behavioral testing methods, data analysis, and critical interpretation.
Collapse
Affiliation(s)
- Jill L. Silverman
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping ServiceNational Institute of Mental HealthBethesdaMarylandUSA
| | - Sarah B. Ethridge
- Neurodevelopmental and Behavioral Phenotyping ServiceNational Institute of Mental HealthBethesdaMarylandUSA
| | - Makayla M. Soller
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Stela P. Petkova
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Ted Abel
- Department of Neuroscience and PharmacologyIowa Neuroscience Institute, University of IowaIowa CityIowaUSA
| | - Melissa D. Bauman
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Edward S. Brodkin
- Department of PsychiatryPerelman School of Medicine at the University of Pennsylvania, Translational Research LaboratoryPhiladelphiaPennsylvaniaUSA
| | - Hala Harony‐Nicolas
- Seaver Autism Center for Research and TreatmentIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Markus Wöhr
- Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological PsychologySocial and Affective Neuroscience Research Group, KU LeuvenLeuvenBelgium,Leuven Brain InstituteKU LeuvenLeuvenBelgium,Faculty of Psychology, Experimental and Biological Psychology, Behavioral NeurosciencePhilipps‐University of MarburgMarburgGermany,Center for Mind, Brain, and BehaviorPhilipps‐University of MarburgMarburgGermany
| | - Alycia Halladay
- Autism Science FoundationUSA,Department of Pharmacology and ToxicologyRutgers UniversityPiscatawayNew JerseyUSA
| |
Collapse
|
63
|
Yu Y, Zhang B, Ji P, Zuo Z, Huang Y, Wang N, Liu C, Liu SJ, Zhao F. Changes to gut amino acid transporters and microbiome associated with increased E/I ratio in Chd8 +/- mouse model of ASD-like behavior. Nat Commun 2022; 13:1151. [PMID: 35241668 PMCID: PMC8894489 DOI: 10.1038/s41467-022-28746-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD), a group of neurodevelopmental disorders characterized by social communication deficits and stereotyped behaviors, may be associated with changes to the gut microbiota. However, how gut commensal bacteria modulate brain function in ASD remains unclear. Here, we used chromodomain helicase DNA-binding protein 8 (CHD8) haploinsufficient mice as a model of ASD to elucidate the pathways through which the host and gut microbiota interact with each other. We found that increased levels of amino acid transporters in the intestines of the mouse model of ASD contribute to the high level of serum glutamine and the increased excitation/inhibition (E/I) ratio in the brain. In addition, elevated α-defensin levels in the haploinsufficient mice resulted in dysregulation of the gut microbiota characterized by a reduced abundance of Bacteroides. Furthermore, supplementation with Bacteroides uniformis improved the ASD-like behaviors and restored the E/I ratio in the brain by decreasing intestinal amino acid transport and the serum glutamine levels. Our study demonstrates associations between changes in the gut microbiota and amino acid transporters, and ASD-like behavioral and electrophysiology phenotypes, in a mouse model.
Collapse
Affiliation(s)
- You Yu
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Bing Zhang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Peifeng Ji
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Zhenqiang Zuo
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Yongxi Huang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Ning Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Chang Liu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
64
|
Terashima H, Minatohara K, Maruoka H, Okabe S. Imaging neural circuit pathology of autism spectrum disorders: autism-associated genes, animal models and the application of in vivo two-photon imaging. Microscopy (Oxf) 2022; 71:i81-i99. [DOI: 10.1093/jmicro/dfab039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/11/2021] [Accepted: 11/08/2021] [Indexed: 11/12/2022] Open
Abstract
Abstract
Recent advances in human genetics identified genetic variants involved in causing autism spectrum disorders (ASDs). Mouse models that mimic mutations found in patients with ASD exhibit behavioral phenotypes consistent with ASD symptoms. These mouse models suggest critical biological factors of ASD etiology. Another important implication of ASD genetics is the enrichment of ASD risk genes in molecules involved in developing synapses and regulating neural circuit function. Sophisticated in vivo imaging technologies applied to ASD mouse models identify common synaptic impairments in the neocortex, with genetic-mutation-specific defects in local neural circuits. In this article, we review synapse- and circuit-level phenotypes identified by in vivo two-photon imaging in multiple mouse models of ASD and discuss the contributions of altered synapse properties and neural circuit activity to ASD pathogenesis.
Collapse
Affiliation(s)
- Hiroshi Terashima
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Keiichiro Minatohara
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hisato Maruoka
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
65
|
Chen X, Chen T, Dong C, Chen H, Dong X, Yang L, Hu L, Wang H, Wu B, Yao Y, Xiong Y, Xiong M, Lin Y, Zhou W. Deletion of CHD8 in cerebellar granule neuron progenitors leads to severe cerebellar hypoplasia, ataxia and psychiatric behavior in mice. J Genet Genomics 2022; 49:859-869. [DOI: 10.1016/j.jgg.2022.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 12/22/2022]
|
66
|
Neural Transcriptomic Analysis of Sex Differences in Autism Spectrum Disorder: Current Insights and Future Directions. Biol Psychiatry 2022; 91:53-60. [PMID: 33551190 DOI: 10.1016/j.biopsych.2020.11.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/18/2022]
Abstract
Autism spectrum disorder (ASD) is consistently diagnosed 3 to 5 times more frequently in males than females, a dramatically sex-biased prevalence that suggests the involvement of sex-differential biological factors in modulating risk. The genomic scale of transcriptomic analyses of human brain tissue can provide an unbiased approach for identifying genes and associated functional processes at the intersection of sex-differential and ASD-impacted neurobiology. Several studies characterizing gene expression changes in the ASD brain have been published in recent years with increasing sample size and cellular resolution. These studies report several convergent patterns across data sets and genetically heterogeneous samples in the ASD brain, including elevated expression of gene sets associated with glial and immune function, and reduced expression of gene sets associated with neuronal and synaptic functions. Assessment of neurotypical cortex tissue has reported parallel patterns by sex, with male-elevated expression of overlapping sets of glial/immune-related genes and female-biased expression of neuron-associated genes, suggesting potential roles for these cell types in sex-differential ASD risk mechanisms. However, validating and further exploring these mechanisms is challenged by the available data, as existing studies of ASD brain include a limited number of female ASD donors and focus predominantly on cortex regions not known to show pronounced sex-differential morphology or function. With this review, we summarize convergent findings from several landmark studies of the transcriptome in ASD brain and their relationship to sex-differential gene expression, and we discuss limitations and remaining questions regarding transcriptomic analysis of sex differences in ASD.
Collapse
|
67
|
|
68
|
Zarantonello G, Arnoldi M, Filosi M, Tebaldi T, Spirito G, Barbieri A, Gustincich S, Sanges R, Domenici E, Di Leva F, Biagioli M. Natural SINEUP RNAs in Autism Spectrum Disorders: RAB11B-AS1 Dysregulation in a Neuronal CHD8 Suppression Model Leads to RAB11B Protein Increase. Front Genet 2021; 12:745229. [PMID: 34880900 PMCID: PMC8647803 DOI: 10.3389/fgene.2021.745229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/20/2021] [Indexed: 11/26/2022] Open
Abstract
CHD8 represents one of the highest confidence genetic risk factors implied in Autism Spectrum Disorders, with most mutations leading to CHD8 haploinsufficiency and the insurgence of specific phenotypes, such as macrocephaly, facial dysmorphisms, intellectual disability, and gastrointestinal complaints. While extensive studies have been conducted on the possible consequences of CHD8 suppression and protein coding RNAs dysregulation during neuronal development, the effects of transcriptional changes of long non-coding RNAs (lncRNAs) remain unclear. In this study, we focused on a peculiar class of natural antisense lncRNAs, SINEUPs, that enhance translation of a target mRNA through the activity of two RNA domains, an embedded transposable element sequence and an antisense region. By looking at dysregulated transcripts following CHD8 knock down (KD), we first identified RAB11B-AS1 as a potential SINEUP RNA for its domain configuration. Then we demonstrated that such lncRNA is able to increase endogenous RAB11B protein amounts without affecting its transcriptional levels. RAB11B has a pivotal role in vesicular trafficking, and mutations on this gene correlate with intellectual disability and microcephaly. Thus, our study discloses an additional layer of molecular regulation which is altered by CHD8 suppression. This represents the first experimental confirmation that naturally occurring SINEUP could be involved in ASD pathogenesis and underscores the importance of dysregulation of functional lncRNAs in neurodevelopment.
Collapse
Affiliation(s)
- Giulia Zarantonello
- Laboratory of Neuroepigenetics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Michele Arnoldi
- Laboratory of Neuroepigenetics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Michele Filosi
- Laboratory of Neurogenomic Biomarkers, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Toma Tebaldi
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, United States.,Laboratory of RNA and Disease Data Science, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Giovanni Spirito
- Laboratory of Computational Genomics, Area of Neuroscience, International School of Advanced Studies (SISSA), Trieste, Italy.,Central RNA Laboratory, Italian Institute of Technology (IIT), Genova, Italy
| | - Anna Barbieri
- Laboratory of Neuroepigenetics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Stefano Gustincich
- Central RNA Laboratory, Italian Institute of Technology (IIT), Genova, Italy
| | - Remo Sanges
- Laboratory of Computational Genomics, Area of Neuroscience, International School of Advanced Studies (SISSA), Trieste, Italy.,Central RNA Laboratory, Italian Institute of Technology (IIT), Genova, Italy
| | - Enrico Domenici
- Laboratory of Neurogenomic Biomarkers, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.,Fondazione The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, Italy
| | - Francesca Di Leva
- Laboratory of Neuroepigenetics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Marta Biagioli
- Laboratory of Neuroepigenetics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| |
Collapse
|
69
|
Markenscoff-Papadimitriou E, Binyameen F, Whalen S, Price J, Lim K, Ypsilanti AR, Catta-Preta R, Pai ELL, Mu X, Xu D, Pollard KS, Nord AS, State MW, Rubenstein JL. Autism risk gene POGZ promotes chromatin accessibility and expression of clustered synaptic genes. Cell Rep 2021; 37:110089. [PMID: 34879283 PMCID: PMC9512081 DOI: 10.1016/j.celrep.2021.110089] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 10/11/2021] [Accepted: 11/11/2021] [Indexed: 12/31/2022] Open
Abstract
Deleterious genetic variants in POGZ, which encodes the chromatin regulator Pogo Transposable Element with ZNF Domain protein, are strongly associated with autism spectrum disorder (ASD). Although it is a high-confidence ASD risk gene, the neurodevelopmental functions of POGZ remain unclear. Here we reveal the genomic binding of POGZ in the developing forebrain at euchromatic loci and gene regulatory elements (REs). We profile chromatin accessibility and gene expression in Pogz-/- mice and show that POGZ promotes the active chromatin state and transcription of clustered synaptic genes. We further demonstrate that POGZ forms a nuclear complex and co-occupies loci with ADNP, another high-confidence ASD risk gene, and provide evidence that POGZ regulates other neurodevelopmental disorder risk genes as well. Our results reveal a neurodevelopmental function of an ASD risk gene and identify molecular targets that may elucidate its function in ASD.
Collapse
Affiliation(s)
- Eirene Markenscoff-Papadimitriou
- Department of Psychiatry, Langley Porter Psychiatric Institute, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA.
| | - Fadya Binyameen
- Department of Psychiatry, Langley Porter Psychiatric Institute, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Sean Whalen
- Gladstone Institutes, San Francisco, CA, USA
| | - James Price
- Department of Psychiatry, Langley Porter Psychiatric Institute, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Kenneth Lim
- Department of Psychiatry, Langley Porter Psychiatric Institute, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Athena R Ypsilanti
- Department of Psychiatry, Langley Porter Psychiatric Institute, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Rinaldo Catta-Preta
- Departments of Neurobiology, Physiology, and Behavior and Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
| | - Emily Ling-Lin Pai
- Department of Psychiatry, Langley Porter Psychiatric Institute, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | | | | | - Katherine S Pollard
- Gladstone Institutes, San Francisco, CA, USA; Chan-Zuckerberg Biohub, San Francisco, CA, USA; Institute for Computational Health Sciences, University of California, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA; Quantitative Biology Institute, University of California, San Francisco, CA, USA
| | - Alex S Nord
- Departments of Neurobiology, Physiology, and Behavior and Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
| | - Matthew W State
- Department of Psychiatry, Langley Porter Psychiatric Institute, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - John L Rubenstein
- Department of Psychiatry, Langley Porter Psychiatric Institute, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA.
| |
Collapse
|
70
|
Chen J, Lambo ME, Ge X, Dearborn JT, Liu Y, McCullough KB, Swift RG, Tabachnick DR, Tian L, Noguchi K, Garbow JR, Constantino JN, Gabel HW, Hengen KB, Maloney SE, Dougherty JD. A MYT1L syndrome mouse model recapitulates patient phenotypes and reveals altered brain development due to disrupted neuronal maturation. Neuron 2021; 109:3775-3792.e14. [PMID: 34614421 PMCID: PMC8668036 DOI: 10.1016/j.neuron.2021.09.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 05/07/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023]
Abstract
Human genetics have defined a new neurodevelopmental syndrome caused by loss-of-function mutations in MYT1L, a transcription factor known for enabling fibroblast-to-neuron conversions. However, how MYT1L mutation causes intellectual disability, autism, ADHD, obesity, and brain anomalies is unknown. Here, we developed a Myt1l haploinsufficient mouse model that develops obesity, white-matter thinning, and microcephaly, mimicking common clinical phenotypes. During brain development we discovered disrupted gene expression, mediated in part by loss of Myt1l gene-target activation, and identified precocious neuronal differentiation as the mechanism for microcephaly. In contrast, in adults we discovered that mutation results in failure of transcriptional and chromatin maturation, echoed in disruptions in baseline physiological properties of neurons. Myt1l haploinsufficiency also results in behavioral anomalies, including hyperactivity, muscle weakness, and social alterations, with more severe phenotypes in males. Overall, our findings provide insight into the mechanistic underpinnings of this disorder and enable future preclinical studies.
Collapse
Affiliation(s)
- Jiayang Chen
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Mary E Lambo
- Department of Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xia Ge
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Joshua T Dearborn
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yating Liu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Katherine B McCullough
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Raylynn G Swift
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Dora R Tabachnick
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Lucy Tian
- Department of Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin Noguchi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Joel R Garbow
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA; Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA; Alvin J Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO USA
| | - John N Constantino
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Harrison W Gabel
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Keith B Hengen
- Department of Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
71
|
Catta-Preta R, Zdilar I, Jenner B, Doisy ET, Tercovich K, Nord AS, Gurkoff GG. Transcriptional Pathology Evolves over Time in Rat Hippocampus after Lateral Fluid Percussion Traumatic Brain Injury. Neurotrauma Rep 2021; 2:512-525. [PMID: 34909768 PMCID: PMC8667199 DOI: 10.1089/neur.2021.0021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Traumatic brain injury (TBI) causes acute and lasting impacts on the brain, driving pathology along anatomical, cellular, and behavioral dimensions. Rodent models offer an opportunity to study the temporal progression of disease from injury to recovery. Transcriptomic and epigenomic analysis were applied to evaluate gene expression in ipsilateral hippocampus at 1 and 14 days after sham (n = 2 and 4, respectively per time point) and moderate lateral fluid percussion injury (n = 4 per time point). This enabled the identification of dynamic changes and differential gene expression (differentially expressed genes; DEGs) modules linked to underlying epigenetic response. We observed acute signatures associated with cell death, astrocytosis, and neurotransmission that largely recovered by 2 weeks. Inflammation and immune signatures segregated into upregulated modules with distinct expression trajectories and functions. Whereas most down-regulated genes recovered by 14 days, two modules with delayed and persistent changes were associated with cholesterol metabolism, amyloid beta clearance, and neurodegeneration. Differential expression was paralleled by changes in histone H3 lysine residue 4 trimethylation at the promoters of DEGs at 1 day post-TBI, with the strongest changes observed for inflammation and immune response genes. These results demonstrate how integrated genomics analysis in the pre-clinical setting has the potential to identify stage-specific biomarkers for injury and/or recovery. Though limited in scope here, our general strategy has the potential to capture pathological signatures over time and evaluate treatment efficacy at the systems level.
Collapse
Affiliation(s)
- Rinaldo Catta-Preta
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, California, USA
- Center for Neuroscience, University of California Davis, Davis, California, USA
| | - Iva Zdilar
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, California, USA
- Center for Neuroscience, University of California Davis, Davis, California, USA
| | - Bradley Jenner
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, California, USA
- Center for Neuroscience, University of California Davis, Davis, California, USA
| | - Emily T. Doisy
- Department of Neurological Surgery, University of California Davis, Davis, California, USA
| | - Kayleen Tercovich
- Department of Neurological Surgery, University of California Davis, Davis, California, USA
- Center for Neuroscience, University of California Davis, Davis, California, USA
| | - Alex S. Nord
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, California, USA
- Center for Neuroscience, University of California Davis, Davis, California, USA
| | - Gene G. Gurkoff
- Department of Neurological Surgery, University of California Davis, Davis, California, USA
- Center for Neuroscience, University of California Davis, Davis, California, USA
| |
Collapse
|
72
|
Wade AA, van den Ameele J, Cheetham SW, Yakob R, Brand AH, Nord AS. In vivo targeted DamID identifies CHD8 genomic targets in fetal mouse brain. iScience 2021; 24:103234. [PMID: 34746699 PMCID: PMC8551073 DOI: 10.1016/j.isci.2021.103234] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/11/2021] [Accepted: 10/04/2021] [Indexed: 01/15/2023] Open
Abstract
Genetic studies of autism have revealed causal roles for chromatin remodeling gene mutations. Chromodomain helicase DNA binding protein 8 (CHD8) encodes a chromatin remodeler with significant de novo mutation rates in sporadic autism. However, relationships between CHD8 genomic function and autism-relevant biology remain poorly elucidated. Published studies utilizing ChIP-seq to map CHD8 protein-DNA interactions have high variability, consistent with technical challenges and limitations associated with this method. Thus, complementary approaches are needed to establish CHD8 genomic targets and regulatory functions in developing brain. We used in utero CHD8 Targeted DamID followed by sequencing (TaDa-seq) to characterize CHD8 binding in embryonic mouse cortex. CHD8 TaDa-seq reproduced interaction patterns observed from ChIP-seq and further highlighted CHD8 distal interactions associated with neuronal loci. This study establishes TaDa-seq as a useful alternative for mapping protein-DNA interactions in vivo and provides insights into the regulatory targets of CHD8 and autism-relevant pathophysiology associated with CHD8 mutations.
Collapse
Affiliation(s)
- A. Ayanna Wade
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA 95616, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, USA
| | - Jelle van den Ameele
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 1TN, UK
| | - Seth W. Cheetham
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 1TN, UK
| | - Rebecca Yakob
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 1TN, UK
| | - Andrea H. Brand
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 1TN, UK
| | - Alex S. Nord
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA 95616, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
73
|
Abstract
Chromatin is highly dynamic, undergoing continuous global changes in its structure and type of histone and DNA modifications governed by processes such as transcription, repair, replication, and recombination. Members of the chromodomain helicase DNA-binding (CHD) family of enzymes are ATP-dependent chromatin remodelers that are intimately involved in the regulation of chromatin dynamics, altering nucleosomal structure and DNA accessibility. Genetic studies in yeast, fruit flies, zebrafish, and mice underscore essential roles of CHD enzymes in regulating cellular fate and identity, as well as proper embryonic development. With the advent of next-generation sequencing, evidence is emerging that these enzymes are subjected to frequent DNA copy number alterations or mutations and show aberrant expression in malignancies and other human diseases. As such, they might prove to be valuable biomarkers or targets for therapeutic intervention.
Collapse
Affiliation(s)
- Andrej Alendar
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066CX, The Netherlands
| | - Anton Berns
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066CX, The Netherlands
| |
Collapse
|
74
|
Ding S, Lan X, Meng Y, Yan C, Li M, Li X, Chen J, Jiang W. CHD8 safeguards early neuroectoderm differentiation in human ESCs and protects from apoptosis during neurogenesis. Cell Death Dis 2021; 12:981. [PMID: 34686651 PMCID: PMC8536677 DOI: 10.1038/s41419-021-04292-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022]
Abstract
The chromatin remodeler CHD8, which belongs to the ATP-dependent chromatin remodelers CHD family, is one of the most high-risk mutated genes in autism spectrum disorders. However, the role of CHD8 in neural differentiation and the mechanism of CHD8 in autism remains unclear, despite there are a few studies based on the CHD8 haploinsufficient models. Here, we generate the CHD8 knockout human ESCs by CRISPR/Cas9 technology and characterize the effect of loss-of-function of CHD8 on pluripotency maintenance and lineage determination by utilizing efficient directed differentiation protocols. The results show loss-of-function of CHD8 does not affect human ESC maintenance although having slight effect on proliferation and cell cycle. Interestingly, CHD8 depletion results in defective neuroectoderm differentiation, along with severe cell death in neural progenitor stage. Transcriptome analysis also indicates CHD8 does not alter the expression of pluripotent genes in ESC stage, but in neural progenitor cells depletion of CHD8 induces the abnormal expression of the apoptosis genes and suppresses neuroectoderm-related genes. These results provide the evidence that CHD8 plays an essential role in the pluripotency exit and neuroectoderm differentiation as well as the regulation of apoptosis during neurogenesis.
Collapse
Affiliation(s)
- Song Ding
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China
| | - Xianchun Lan
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China
| | - Yajing Meng
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China
| | - Chenchao Yan
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China
| | - Mao Li
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China
| | - Xiang Li
- Department of Neurosurgery, China Brain Research Center, Medical Research Institute, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China.
| | - Jian Chen
- Chinese Institute for Brain Research (Beijing), Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, 102206, Beijing, China.
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, 215123, Suzhou, China.
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China.
- Human Genetics Resource Preservation Center of Wuhan University, 430071, Wuhan, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, 430071, Wuhan, China.
| |
Collapse
|
75
|
Berg EL, Petkova SP, Born HA, Adhikari A, Anderson AE, Silverman JL. Insulin-like growth factor-2 does not improve behavioral deficits in mouse and rat models of Angelman Syndrome. Mol Autism 2021; 12:59. [PMID: 34526125 PMCID: PMC8444390 DOI: 10.1186/s13229-021-00467-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Angelman Syndrome (AS) is a rare neurodevelopmental disorder for which there is currently no cure or effective therapeutic. Since the genetic cause of AS is known to be dysfunctional expression of the maternal allele of ubiquitin protein ligase E3A (UBE3A), several genetic animal models of AS have been developed. Both the Ube3a maternal deletion mouse and rat models of AS reliably demonstrate behavioral phenotypes of relevance to AS and therefore offer suitable in vivo systems in which to test potential therapeutics. One promising candidate treatment is insulin-like growth factor-2 (IGF-2), which has recently been shown to ameliorate behavioral deficits in the mouse model of AS and improve cognitive abilities across model systems. METHODS We used both the Ube3a maternal deletion mouse and rat models of AS to evaluate the ability of IGF-2 to improve electrophysiological and behavioral outcomes. RESULTS Acute systemic administration of IGF-2 had an effect on electrophysiological activity in the brain and on a metric of motor ability; however the effects were not enduring or extensive. Additional metrics of motor behavior, learning, ambulation, and coordination were unaffected and IGF-2 did not improve social communication, seizure threshold, or cognition. LIMITATIONS The generalizability of these results to humans is difficult to predict and it remains possible that dosing schemes (i.e., chronic or subchronic dosing), routes, and/or post-treatment intervals other than that used herein may show more efficacy. CONCLUSIONS Despite a few observed effects of IGF-2, our results taken together indicate that IGF-2 treatment does not profoundly improve behavioral deficits in mouse or rat models of AS. These findings shed cautionary light on the potential utility of acute systemic IGF-2 administration in the treatment of AS.
Collapse
Affiliation(s)
- Elizabeth L. Berg
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA USA
| | - Stela P. Petkova
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA USA
| | - Heather A. Born
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX USA
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Anna Adhikari
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA USA
| | - Anne E. Anderson
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX USA
| | - Jill L. Silverman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA USA
| |
Collapse
|
76
|
Davies FCJ, Hope JE, McLachlan F, Marshall GF, Kaminioti-Dumont L, Qarkaxhija V, Nunez F, Dando O, Smith C, Wood E, MacDonald J, Hardt O, Abbott CM. Recapitulation of the EEF1A2 D252H neurodevelopmental disorder-causing missense mutation in mice reveals a toxic gain of function. Hum Mol Genet 2021; 29:1592-1606. [PMID: 32160274 DOI: 10.1093/hmg/ddaa042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/24/2022] Open
Abstract
Heterozygous de novo mutations in EEF1A2, encoding the tissue-specific translation elongation factor eEF1A2, have been shown to cause neurodevelopmental disorders including often severe epilepsy and intellectual disability. The mutational profile is unusual; ~50 different missense mutations have been identified but no obvious loss of function mutations, though large heterozygous deletions are known to be compatible with life. A key question is whether the heterozygous missense mutations operate through haploinsufficiency or a gain of function mechanism, an important prerequisite for design of therapeutic strategies. In order both to address this question and to provide a novel model for neurodevelopmental disorders resulting from mutations in EEF1A2, we created a new mouse model of the D252H mutation. This mutation causes the eEF1A2 protein to be expressed at lower levels in brain but higher in muscle in the mice. We compared both heterozygous and homozygous D252H and null mutant mice using behavioural and motor phenotyping alongside molecular modelling and analysis of binding partners. Although the proteomic analysis pointed to a loss of function for the D252H mutant protein, the D252H homozygous mice were more severely affected than null homozygotes on the same genetic background. Mice that are heterozygous for the missense mutation show no behavioural abnormalities but do have sex-specific deficits in body mass and motor function. The phenotyping of our novel mouse lines, together with analysis of molecular modelling and interacting proteins, suggest that the D252H mutation results in a gain of function.
Collapse
Affiliation(s)
- Faith C J Davies
- Centre for Genomic & Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Jilly E Hope
- Centre for Genomic & Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Fiona McLachlan
- Centre for Genomic & Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Grant F Marshall
- Centre for Genomic & Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Laura Kaminioti-Dumont
- Centre for Genomic & Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Vesa Qarkaxhija
- Centre for Genomic & Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Francis Nunez
- Centre for Genomic & Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Owen Dando
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Colin Smith
- Academic Department of Neuropathology, Centre for Clinical Brain Sciences, Edinburgh, EH16 4SB, United Kingdom
| | - Emma Wood
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Josephine MacDonald
- Centre for Genomic & Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Oliver Hardt
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom.,Department of Psychology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Catherine M Abbott
- Centre for Genomic & Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| |
Collapse
|
77
|
Kawamura A, Katayama Y, Nishiyama M, Shoji H, Tokuoka K, Ueta Y, Miyata M, Isa T, Miyakawa T, Hayashi-Takagi A, Nakayama KI. Oligodendrocyte dysfunction due to Chd8 mutation gives rise to behavioral deficits in mice. Hum Mol Genet 2021; 29:1274-1291. [PMID: 32142125 DOI: 10.1093/hmg/ddaa036] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/23/2020] [Accepted: 02/27/2020] [Indexed: 01/01/2023] Open
Abstract
Mutations in the gene encoding the chromatin remodeler CHD8 are strongly associated with autism spectrum disorder (ASD). CHD8 haploinsufficiency also results in autistic phenotypes in humans and mice. Although myelination defects have been observed in individuals with ASD, whether oligodendrocyte dysfunction is responsible for autistic phenotypes has remained unknown. Here we show that reduced expression of CHD8 in oligodendrocytes gives rise to abnormal behavioral phenotypes in mice. CHD8 was found to regulate the expression of many myelination-related genes and to be required for oligodendrocyte maturation and myelination. Ablation of Chd8 specifically in oligodendrocytes of mice impaired myelination, slowed action potential propagation and resulted in behavioral deficits including increased social interaction and anxiety-like behavior, with similar effects being apparent in Chd8 heterozygous mutant mice. Our results thus indicate that CHD8 is essential for myelination and that dysfunction of oligodendrocytes as a result of CHD8 haploinsufficiency gives rise to several neuropsychiatric phenotypes.
Collapse
Affiliation(s)
- Atsuki Kawamura
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuta Katayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Masaaki Nishiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Kota Tokuoka
- Department of Neuroscience, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Yoshifumi Ueta
- Department of Physiology I (Neurophysiology), Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Mariko Miyata
- Department of Physiology I (Neurophysiology), Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Tadashi Isa
- Department of Neuroscience, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Akiko Hayashi-Takagi
- Laboratory of Medical Neuroscience, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma 371-8512, Japan.,PRESTO, Japan Science and Technology Agency (JST), Kawaguchi, Saitama 332-0012, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
78
|
Weissberg O, Elliott E. The Mechanisms of CHD8 in Neurodevelopment and Autism Spectrum Disorders. Genes (Basel) 2021; 12:genes12081133. [PMID: 34440307 PMCID: PMC8393912 DOI: 10.3390/genes12081133] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 11/16/2022] Open
Abstract
Chromodomain-helicase-DNA-binding protein 8 (CHD8) has been identified as one of the genes with the strongest association with autism. The CHD8 protein is a transcriptional regulator that is expressed in nearly all cell types and has been implicated in multiple cellular processes, including cell cycle, cell adhesion, neuronal development, myelination, and synaptogenesis. Considering the central role of CHD8 in the genetics of autism, a deeper understanding of the physiological functions of CHD8 is important to understand the development of the autism phenotype and potential therapeutic targets. Different CHD8 mutant mouse models were developed to determine autism-like phenotypes and to fully understand their mechanisms. Here, we review the current knowledge on CHD8, with an emphasis on mechanistic lessons gained from animal models that have been studied.
Collapse
|
79
|
D'Souza L, Channakkar AS, Muralidharan B. Chromatin remodelling complexes in cerebral cortex development and neurodevelopmental disorders. Neurochem Int 2021; 147:105055. [PMID: 33964373 PMCID: PMC7611358 DOI: 10.1016/j.neuint.2021.105055] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/11/2021] [Accepted: 04/24/2021] [Indexed: 12/19/2022]
Abstract
The diverse number of neurons in the cerebral cortex are generated during development by neural stem cells lining the ventricle, and they continue maturing postnatally. Dynamic chromatin regulation in these neural stem cells is a fundamental determinant of the emerging property of the functional neural network, and the chromatin remodellers are critical determinants of this process. Chromatin remodellers participate in several steps of this process from proliferation, differentiation, migration leading to complex network formation which forms the basis of higher-order functions of cognition and behaviour. Here we review the role of these ATP-dependent chromatin remodellers in cortical development in health and disease and highlight several key mouse mutants of the subunits of the complexes which have revealed how the remodelling mechanisms control the cortical stem cell chromatin landscape for expression of stage-specific transcripts. Consistent with their role in cortical development, several putative risk variants in the subunits of the remodelling complexes have been identified as the underlying causes of several neurodevelopmental disorders. A basic understanding of the detailed molecular mechanism of their action is key to understating how mutations in the same networks lead to disease pathologies and perhaps pave the way for therapeutic development for these complex multifactorial disorders.
Collapse
Affiliation(s)
- Leora D'Souza
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India
| | - Asha S Channakkar
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India
| | - Bhavana Muralidharan
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India.
| |
Collapse
|
80
|
Amar M, Pramod AB, Yu NK, Herrera VM, Qiu LR, Moran-Losada P, Zhang P, Trujillo CA, Ellegood J, Urresti J, Chau K, Diedrich J, Chen J, Gutierrez J, Sebat J, Ramanathan D, Lerch JP, Yates JR, Muotri AR, Iakoucheva LM. Autism-linked Cullin3 germline haploinsufficiency impacts cytoskeletal dynamics and cortical neurogenesis through RhoA signaling. Mol Psychiatry 2021; 26:3586-3613. [PMID: 33727673 PMCID: PMC8443683 DOI: 10.1038/s41380-021-01052-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 01/12/2021] [Accepted: 02/12/2021] [Indexed: 01/01/2023]
Abstract
E3-ubiquitin ligase Cullin3 (Cul3) is a high confidence risk gene for autism spectrum disorder (ASD) and developmental delay (DD). To investigate how Cul3 mutations impact brain development, we generated a haploinsufficient Cul3 mouse model using CRISPR/Cas9 genome engineering. Cul3 mutant mice exhibited social and cognitive deficits and hyperactive behavior. Brain MRI found decreased volume of cortical regions and changes in many other brain regions of Cul3 mutant mice starting from early postnatal development. Spatiotemporal transcriptomic and proteomic profiling of embryonic, early postnatal and adult brain implicated neurogenesis and cytoskeletal defects as key drivers of Cul3 functional impact. Specifically, dendritic growth, filamentous actin puncta, and spontaneous network activity were reduced in Cul3 mutant mice. Inhibition of small GTPase RhoA, a molecular substrate of Cul3 ligase, rescued dendrite length and network activity phenotypes. Our study identified defects in neuronal cytoskeleton and Rho signaling as the primary targets of Cul3 mutation during brain development.
Collapse
Affiliation(s)
- Megha Amar
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Akula Bala Pramod
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Nam-Kyung Yu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Lily R Qiu
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, ON, Canada
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neuroscience, The University of Oxford, Oxford, UK
| | | | - Pan Zhang
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Cleber A Trujillo
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics/Rady Children's Hospital San Diego, University of California San Diego, La Jolla, CA, USA
| | - Jacob Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, ON, Canada
| | - Jorge Urresti
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Kevin Chau
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Jolene Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Jiaye Chen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Jessica Gutierrez
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Jonathan Sebat
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Beyster Center for Psychiatric Genomics, University of California San Diego, La Jolla, CA, USA
| | - Dhakshin Ramanathan
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Jason P Lerch
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, ON, Canada
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neuroscience, The University of Oxford, Oxford, UK
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Alysson R Muotri
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Pediatrics/Rady Children's Hospital San Diego, University of California San Diego, La Jolla, CA, USA.
- Kavli Institute for Brain and Mind, University of California San Diego, La Jolla, CA, USA.
- Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA, USA.
| | - Lilia M Iakoucheva
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
81
|
Ellingford RA, Panasiuk MJ, de Meritens ER, Shaunak R, Naybour L, Browne L, Basson MA, Andreae LC. Cell-type-specific synaptic imbalance and disrupted homeostatic plasticity in cortical circuits of ASD-associated Chd8 haploinsufficient mice. Mol Psychiatry 2021; 26:3614-3624. [PMID: 33837267 PMCID: PMC8505247 DOI: 10.1038/s41380-021-01070-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 02/28/2021] [Accepted: 03/19/2021] [Indexed: 12/11/2022]
Abstract
Heterozygous mutation of chromodomain helicase DNA binding protein 8 (CHD8) is strongly associated with autism spectrum disorder (ASD) and results in dysregulated expression of neurodevelopmental and synaptic genes during brain development. To reveal how these changes affect ASD-associated cortical circuits, we studied synaptic transmission in the prefrontal cortex of a haploinsufficient Chd8 mouse model. We report profound alterations to both excitatory and inhibitory synaptic transmission onto deep layer projection neurons, resulting in a reduced excitatory:inhibitory balance, which were found to vary dynamically across neurodevelopment and result from distinct effects of reduced Chd8 expression within individual neuronal subtypes. These changes were associated with disrupted regulation of homeostatic plasticity mechanisms operating via spontaneous neurotransmission. These findings therefore directly implicate CHD8 mutation in the disruption of ASD-relevant circuits in the cortex.
Collapse
Affiliation(s)
- Robert A Ellingford
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Centre for Craniofacial & Regenerative Biology, King's College London, London, UK
| | - Martyna J Panasiuk
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Emilie Rabesahala de Meritens
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Raghav Shaunak
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Liam Naybour
- Centre for Craniofacial & Regenerative Biology, King's College London, London, UK
| | - Lorcan Browne
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - M Albert Basson
- Centre for Craniofacial & Regenerative Biology, King's College London, London, UK.
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| | - Laura C Andreae
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
82
|
Falcone C, Mevises NY, Hong T, Dufour B, Chen X, Noctor SC, Martínez Cerdeño V. Neuronal and glial cell number is altered in a cortical layer-specific manner in autism. AUTISM : THE INTERNATIONAL JOURNAL OF RESEARCH AND PRACTICE 2021; 25:2238-2253. [PMID: 34107793 DOI: 10.1177/13623613211014408] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
LAY ABSTRACT The cerebral cortex affected with autism spectrum disorder presents changes in the number of neurons and glia cells, possibly leading to a dysregulation of brain circuits and affecting behavior. However, little is known about cell number alteration in specific layers of the cortex in autism spectrum disorder. We found an increase in the number of neurons and a decrease in the number of astrocytes in specific layers of the prefrontal cortex in postmortem human brains from autism spectrum disorder cases. We hypothesize that this may be due to a failure in neural stem cells to shift differentiation from neurons to glial cells during prenatal brain development. These data provide key anatomical findings that contribute to the bases of autism spectrum disorder pathogenesis.
Collapse
Affiliation(s)
- Carmen Falcone
- UC Davis School of Medicine, USA.,Institute for Pediatric Regenerative Medicine, and Shriners Hospitals for Children of Northern California, USA
| | - Natalie-Ya Mevises
- UC Davis School of Medicine, USA.,Institute for Pediatric Regenerative Medicine, and Shriners Hospitals for Children of Northern California, USA
| | - Tiffany Hong
- UC Davis School of Medicine, USA.,Institute for Pediatric Regenerative Medicine, and Shriners Hospitals for Children of Northern California, USA
| | - Brett Dufour
- UC Davis School of Medicine, USA.,Institute for Pediatric Regenerative Medicine, and Shriners Hospitals for Children of Northern California, USA
| | - Xiaohui Chen
- UC Davis School of Medicine, USA.,Institute for Pediatric Regenerative Medicine, and Shriners Hospitals for Children of Northern California, USA
| | | | - Verónica Martínez Cerdeño
- UC Davis School of Medicine, USA.,Institute for Pediatric Regenerative Medicine, and Shriners Hospitals for Children of Northern California, USA
| |
Collapse
|
83
|
Ahmed R, Sarwar S, Hu J, Cardin V, Qiu LR, Zapata G, Vandeleur L, Yan K, Lerch JP, Corbett MA, Gecz J, Picketts DJ. Transgenic mice with an R342X mutation in Phf6 display clinical features of Börjeson-Forssman-Lehmann Syndrome. Hum Mol Genet 2021; 30:575-594. [PMID: 33772537 PMCID: PMC8120135 DOI: 10.1093/hmg/ddab081] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/24/2021] [Accepted: 03/16/2021] [Indexed: 12/26/2022] Open
Abstract
The PHF6 mutation c.1024C > T; p.R342X, is a recurrent cause of Börjeson-Forssman-Lehmann Syndrome (BFLS), a neurodevelopmental disorder characterized by moderate-severe intellectual disability, truncal obesity, gynecomastia, hypogonadism, long tapering fingers and large ears (MIM#301900). Here, we generated transgenic mice with the identical substitution (R342X mice) using CRISPR technology. We show that the p.R342X mutation causes a reduction in PHF6 protein levels, in both human and mice, from nonsense-mediated decay and nonsense-associated alternative splicing, respectively. Magnetic resonance imaging studies indicated that R342X mice had a reduced brain volume on a mixed genetic background but developed hydrocephaly and a high incidence of postnatal death on a C57BL/6 background. Cortical development proceeded normally, while hippocampus and hypothalamus relative brain volumes were altered. A hypoplastic anterior pituitary was also observed that likely contributes to the small size of the R342X mice. Behavior testing demonstrated deficits in associative learning, spatial memory and an anxiolytic phenotype. Taken together, the R342X mice represent a good preclinical model of BFLS that will allow further dissection of PHF6 function and disease pathogenesis.
Collapse
Affiliation(s)
- Raies Ahmed
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
- Departments of Biochemistry, Microbiology, & Immunology, Ottawa, Ontario K1H 8M5, Canada
| | - Shihab Sarwar
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
| | - Jinghua Hu
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
| | - Valérie Cardin
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
- Cellular & Molecular Medicine, Ottawa, Ontario K1H 8M5, Canada
| | - Lily R Qiu
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario M5T 3H7, Canada
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Gerardo Zapata
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
- Departments of Biochemistry, Microbiology, & Immunology, Ottawa, Ontario K1H 8M5, Canada
| | - Lucianne Vandeleur
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Keqin Yan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario M5T 3H7, Canada
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Mark A Corbett
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Jozef Gecz
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - David J Picketts
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
- Departments of Biochemistry, Microbiology, & Immunology, Ottawa, Ontario K1H 8M5, Canada
- Cellular & Molecular Medicine, Ottawa, Ontario K1H 8M5, Canada
- Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
84
|
Oxytocin ameliorates impaired social behavior in a Chd8 haploinsufficiency mouse model of autism. BMC Neurosci 2021; 22:32. [PMID: 33933000 PMCID: PMC8088024 DOI: 10.1186/s12868-021-00631-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 03/24/2021] [Indexed: 11/18/2022] Open
Abstract
Background Autism spectrum disorder (ASD) is characterized by the core symptoms of impaired social interactions. Increasing evidence suggests that ASD has a strong genetic link with mutations in chromodomain helicase DNA binding protein 8 (CHD8), a gene encoding a chromatin remodeler. It has previously been shown that Chd8 haplodeficient male mice manifest ASD-like behavioral characteristics such as anxiety and altered social behavior. Along with that, oxytocin (OT) is one of the main neuropeptides involved in social behavior. Administration of OT has shown improvement of social behavior in genetic animal models of ASD. The present study was undertaken to further explore behavioral abnormalities of Chd8 haplodeficient mice of both sexes, their link with OT, and possible effects of OT administration. First, we performed a battery of behavioral tests on wild-type and Chd8+/∆SL female and male mice. Next, we measured plasma OT levels and finally studied the effects of intraperitoneal OT injection on observed behavioral deficits. Results We showed general anxiety phenotype in Chd8+/∆SL mice regardless of sex, the depressive phenotype in Chd8+/∆SL female mice only and bidirectional social deficit in female and male mice. We observed decreased level of OT in Chd+/∆SL mice, possibly driven by males. Mice injected by OT demonstrated recovery of social behavior, while reduced anxiety was observed only in male mice. Conclusions Here, we demonstrated that abnormal social behaviors were observed in both male and female Chd8+/∆SL mice. The ability of peripheral OT administration to affect such behaviors along with altered plasma OT levels indicated a possible link between Chd8 + /∆SL and OT in the pathogenesis of ASD as well as the possible usefulness of OT as a therapeutic tool for ASD patients with CHD8 mutations. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-021-00631-6.
Collapse
|
85
|
Cederquist GY, Tchieu J, Callahan SJ, Ramnarine K, Ryan S, Zhang C, Rittenhouse C, Zeltner N, Chung SY, Zhou T, Chen S, Betel D, White RM, Tomishima M, Studer L. A Multiplex Human Pluripotent Stem Cell Platform Defines Molecular and Functional Subclasses of Autism-Related Genes. Cell Stem Cell 2021; 27:35-49.e6. [PMID: 32619517 DOI: 10.1016/j.stem.2020.06.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/26/2020] [Accepted: 06/05/2020] [Indexed: 01/12/2023]
Abstract
Autism is a clinically heterogeneous neurodevelopmental disorder characterized by impaired social interactions, restricted interests, and repetitive behaviors. Despite significant advances in the genetics of autism, understanding how genetic changes perturb brain development and affect clinical symptoms remains elusive. Here, we present a multiplex human pluripotent stem cell (hPSC) platform, in which 30 isogenic disease lines are pooled in a single dish and differentiated into prefrontal cortex (PFC) lineages to efficiently test early-developmental hypotheses of autism. We define subgroups of autism mutations that perturb PFC neurogenesis and are correlated to abnormal WNT/βcatenin responses. Class 1 mutations (8 of 27) inhibit while class 2 mutations (5 of 27) enhance PFC neurogenesis. Remarkably, autism patient data reveal that individuals carrying subclass-specific mutations differ clinically in their corresponding language acquisition profiles. Our study provides a framework to disentangle genetic heterogeneity associated with autism and points toward converging molecular and developmental pathways of diverse autism-associated mutations.
Collapse
Affiliation(s)
- Gustav Y Cederquist
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Weill-Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | - Jason Tchieu
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Scott J Callahan
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Cancer Genetics and Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Gerstner Graduate School of Biomedical Sciences, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Kiran Ramnarine
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Sean Ryan
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Chao Zhang
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA; Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Chelsea Rittenhouse
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Nadja Zeltner
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Center for Molecular Medicine, Department of Cellular Biology, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Sun Young Chung
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Ting Zhou
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Doron Betel
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA; Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Richard M White
- Cancer Genetics and Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Mark Tomishima
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA.
| |
Collapse
|
86
|
Haigh JL, Adhikari A, Copping NA, Stradleigh T, Wade AA, Catta-Preta R, Su-Feher L, Zdilar I, Morse S, Fenton TA, Nguyen A, Quintero D, Agezew S, Sramek M, Kreun EJ, Carter J, Gompers A, Lambert JT, Canales CP, Pennacchio LA, Visel A, Dickel DE, Silverman JL, Nord AS. Deletion of a non-canonical regulatory sequence causes loss of Scn1a expression and epileptic phenotypes in mice. Genome Med 2021; 13:69. [PMID: 33910599 PMCID: PMC8080386 DOI: 10.1186/s13073-021-00884-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/06/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Genes with multiple co-active promoters appear common in brain, yet little is known about functional requirements for these potentially redundant genomic regulatory elements. SCN1A, which encodes the NaV1.1 sodium channel alpha subunit, is one such gene with two co-active promoters. Mutations in SCN1A are associated with epilepsy, including Dravet syndrome (DS). The majority of DS patients harbor coding mutations causing SCN1A haploinsufficiency; however, putative causal non-coding promoter mutations have been identified. METHODS To determine the functional role of one of these potentially redundant Scn1a promoters, we focused on the non-coding Scn1a 1b regulatory region, previously described as a non-canonical alternative transcriptional start site. We generated a transgenic mouse line with deletion of the extended evolutionarily conserved 1b non-coding interval and characterized changes in gene and protein expression, and assessed seizure activity and alterations in behavior. RESULTS Mice harboring a deletion of the 1b non-coding interval exhibited surprisingly severe reductions of Scn1a and NaV1.1 expression throughout the brain. This was accompanied by electroencephalographic and thermal-evoked seizures, and behavioral deficits. CONCLUSIONS This work contributes to functional dissection of the regulatory wiring of a major epilepsy risk gene, SCN1A. We identified the 1b region as a critical disease-relevant regulatory element and provide evidence that non-canonical and seemingly redundant promoters can have essential function.
Collapse
Affiliation(s)
- Jessica L Haigh
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Anna Adhikari
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- MIND Institute and Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, CA, USA
| | - Nycole A Copping
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- MIND Institute and Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, CA, USA
| | - Tyler Stradleigh
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - A Ayanna Wade
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Rinaldo Catta-Preta
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Linda Su-Feher
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Iva Zdilar
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Sarah Morse
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Timothy A Fenton
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- MIND Institute and Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, CA, USA
| | - Anh Nguyen
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Diana Quintero
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Samrawit Agezew
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Michael Sramek
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Ellie J Kreun
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Jasmine Carter
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Andrea Gompers
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Jason T Lambert
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Cesar P Canales
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Len A Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, Berkeley, CA, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, Berkeley, CA, USA
- School of Natural Sciences, University of California, Merced, CA, USA
| | - Diane E Dickel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, Berkeley, CA, USA
| | - Jill L Silverman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA.
- MIND Institute and Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, CA, USA.
| | - Alex S Nord
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA.
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
87
|
Wickramasekara RN, Robertson B, Hulen J, Hallgren J, Stessman HAF. Differential effects by sex with Kmt5b loss. Autism Res 2021; 14:1554-1571. [PMID: 33871180 DOI: 10.1002/aur.2516] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/26/2021] [Accepted: 04/01/2021] [Indexed: 01/21/2023]
Abstract
Lysine methyl transferase 5B (KMT5B) has been recently highlighted as a risk gene in genetic studies of neurodevelopmental disorders (NDDs), specifically, autism spectrum disorder (ASD) and intellectual disability (ID); yet, its role in the brain is not known. The goal of this work was to neurodevelopmentally characterize the effect(s) of KMT5B haploinsufficiency using a mouse model. A Kmt5b gene-trap mouse line was obtained from the Knockout Mouse Project. Wild type (WT) and heterozygous (HET) mice were subjected to a comprehensive neurodevelopmental test battery to assess reflexes, motor behavior, learning/memory, social behavior, repetitive movement, and common ASD comorbidities (obsessive compulsion, depression, and anxiety). Given the strong sex bias observed in the ASD patient population, we tested both a male and female cohort of animals and compared differences between genotypes and sexes. HET mice were significantly smaller than WT littermates starting at postnatal day 10 through young adulthood which was correlated with smaller brain size (i.e., microcephaly). This was more severe in males than females. HET male neonates also had delayed eye opening and significantly weaker reflexes than WT littermates. In young adults, significant differences between genotypes relative to anxiety, depression, fear, and extinction learning were observed. Interestingly, several sexually dimorphic differences were noted including increased repetitive grooming behavior in HET females and an increased latency to hot plate response in HET females versus a decreased latency in HET males. LAY SUMMARY: Lysine methyl transferase 5B (KMT5B) has been recently highlighted as a risk gene in neurodevelopmental disorders (NDDs), specifically, autism spectrum disorder (ASD) and intellectual disability (ID); yet its role in the brain is not known. Our study indicates that mice lacking one genomic copy of Kmt5b show deficits in neonatal reflexes, sociability, repetitive stress-induced grooming, changes in thermal pain sensing, decreased depression and anxiety, increased fear, slower extinction learning, and lower body weight, length, and brain size. Furthermore, several outcomes differed by sex, perhaps mirroring the sex bias in ASD.
Collapse
Affiliation(s)
- Rochelle N Wickramasekara
- Department of Pharmacology & Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska, USA
| | - Brynn Robertson
- Department of Pharmacology & Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska, USA
| | - Jason Hulen
- Department of Pharmacology & Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska, USA
| | - Jodi Hallgren
- Department of Pharmacology & Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska, USA
| | - Holly A F Stessman
- Department of Pharmacology & Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska, USA
| |
Collapse
|
88
|
Adhikari A, Copping NA, Beegle J, Cameron DL, Deng P, O'Geen H, Segal DJ, Fink KD, Silverman JL, Anderson JS. Functional rescue in an Angelman syndrome model following treatment with lentivector transduced hematopoietic stem cells. Hum Mol Genet 2021; 30:1067-1083. [PMID: 33856035 PMCID: PMC8188406 DOI: 10.1093/hmg/ddab104] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by impaired communication skills, ataxia, motor and balance deficits, intellectual disabilities, and seizures. The genetic cause of AS is the neuronal loss of UBE3A expression in the brain. A novel approach, described here, is a stem cell gene therapy which uses lentivector-transduced hematopoietic stem and progenitor cells to deliver functional UBE3A to affected cells. We have demonstrated both the prevention and reversal of AS phenotypes upon transplantation and engraftment of human CD34+ cells transduced with a Ube3a lentivector in a novel immunodeficient Ube3amat−/pat+ IL2rg−/y mouse model of AS. A significant improvement in motor and cognitive behavioral assays as well as normalized delta power measured by electroencephalogram was observed in neonates and adults transplanted with the gene modified cells. Human hematopoietic profiles observed in the lymphoid organs by detection of human immune cells were normal. Expression of UBE3A was detected in the brains of the adult treatment group following immunohistochemical staining illustrating engraftment of the gene-modified cells expressing UBE3A in the brain. As demonstrated with our data, this stem cell gene therapy approach offers a promising treatment strategy for AS, not requiring a critical treatment window.
Collapse
Affiliation(s)
- Anna Adhikari
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Nycole A Copping
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Julie Beegle
- Stem Cell Program, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - David L Cameron
- Stem Cell Program, Department of Neurology, Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Peter Deng
- Stem Cell Program, Department of Neurology, Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Henriette O'Geen
- Department of Biochemistry and Medical Microbiology, UC Davis Genome Center, University of California Davis School of Medicine, Davis, CA 95616, USA
| | - David J Segal
- Department of Biochemistry and Medical Microbiology, UC Davis Genome Center, University of California Davis School of Medicine, Davis, CA 95616, USA
| | - Kyle D Fink
- Stem Cell Program, Department of Neurology, Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jill L Silverman
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Joseph S Anderson
- Stem Cell Program, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
89
|
Cerase A, Young AN, Ruiz NB, Buness A, Sant GM, Arnold M, Di Giacomo M, Ascolani M, Kumar M, Hierholzer A, Trigiante G, Marzi SJ, Avner P. Chd8 regulates X chromosome inactivation in mouse through fine-tuning control of Xist expression. Commun Biol 2021; 4:485. [PMID: 33859315 PMCID: PMC8050208 DOI: 10.1038/s42003-021-01945-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 02/25/2021] [Indexed: 01/22/2023] Open
Abstract
Female mammals achieve dosage compensation by inactivating one of their two X chromosomes during development, a process entirely dependent on Xist, an X-linked long non-coding RNA (lncRNA). At the onset of X chromosome inactivation (XCI), Xist is up-regulated and spreads along the future inactive X chromosome. Contextually, it recruits repressive histone and DNA modifiers that transcriptionally silence the X chromosome. Xist regulation is tightly coupled to differentiation and its expression is under the control of both pluripotency and epigenetic factors. Recent evidence has suggested that chromatin remodelers accumulate at the X Inactivation Center (XIC) and here we demonstrate a new role for Chd8 in Xist regulation in differentiating ES cells, linked to its control and prevention of spurious transcription factor interactions occurring within Xist regulatory regions. Our findings have a broader relevance, in the context of complex, developmentally-regulated gene expression.
Collapse
Affiliation(s)
- Andrea Cerase
- EMBL-Rome, Epigenetics and Neurobiology Unit, Monterotondo, Italy.
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Alexander N Young
- EMBL-Rome, Epigenetics and Neurobiology Unit, Monterotondo, Italy
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nerea Blanes Ruiz
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Andreas Buness
- EMBL-Rome, Epigenetics and Neurobiology Unit, Monterotondo, Italy
- Core Unit for Bioinformatics Data Analysis Universitätsklinikum Bonn, Bonn, Germany
| | - Gabrielle M Sant
- EMBL-Rome, Epigenetics and Neurobiology Unit, Monterotondo, Italy
- Institute of Molecular Biology gGmbH (IMB), Mainz, Germany
| | - Mirjam Arnold
- EMBL-Rome, Epigenetics and Neurobiology Unit, Monterotondo, Italy
- Max Planck Institute for Molecular Genetics, Otto Warburg Laboratory, Berlin, Germany
| | | | - Michela Ascolani
- EMBL-Rome, Epigenetics and Neurobiology Unit, Monterotondo, Italy
| | - Manish Kumar
- EMBL-Rome, Epigenetics and Neurobiology Unit, Monterotondo, Italy
- Department of Allied Health Science, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE, Vijaypura, Karnataka, India
| | - Andreas Hierholzer
- EMBL-Rome, Epigenetics and Neurobiology Unit, Monterotondo, Italy
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Giuseppe Trigiante
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sarah J Marzi
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Philip Avner
- EMBL-Rome, Epigenetics and Neurobiology Unit, Monterotondo, Italy.
| |
Collapse
|
90
|
Kawamura A, Katayama Y, Kakegawa W, Ino D, Nishiyama M, Yuzaki M, Nakayama KI. The autism-associated protein CHD8 is required for cerebellar development and motor function. Cell Rep 2021; 35:108932. [PMID: 33826902 DOI: 10.1016/j.celrep.2021.108932] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 12/24/2020] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
Mutations in the gene encoding the chromatin remodeler chromodomain helicase DNA-binding protein 8 (CHD8) are a highly penetrant risk factor for autism spectrum disorder (ASD). Although cerebellar abnormalities have long been thought to be related to ASD pathogenesis, it has remained largely unknown whether dysfunction of CHD8 in the cerebellum contributes to ASD phenotypes. We here show that cerebellar granule neuron progenitor (GNP)-specific deletion of Chd8 in mice impairs the proliferation and differentiation of these cells as well as gives rise to cerebellar hypoplasia and a motor coordination defect, but not to ASD-like behavioral abnormalities. CHD8 is found to regulate the expression of neuronal genes in GNPs. It also binds preferentially to promoter regions and modulates local chromatin accessibility of transcriptionally active genes in these cells. Our results have thus uncovered a key role for CHD8 in cerebellar development, with important implications for understanding the contribution of this brain region to ASD pathogenesis.
Collapse
Affiliation(s)
- Atsuki Kawamura
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Department of Histology and Cell Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Yuta Katayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.
| | - Wataru Kakegawa
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Daisuke Ino
- Department of Histology and Cell Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Masaaki Nishiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Department of Histology and Cell Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
91
|
Ellegood J, Petkova SP, Kinman A, Qiu LR, Adhikari A, Wade AA, Fernandes D, Lindenmaier Z, Creighton A, Nutter LMJ, Nord AS, Silverman JL, Lerch JP. Neuroanatomy and behavior in mice with a haploinsufficiency of AT-rich interactive domain 1B (ARID1B) throughout development. Mol Autism 2021; 12:25. [PMID: 33757588 PMCID: PMC7986278 DOI: 10.1186/s13229-021-00432-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND One of the causal mechanisms underlying neurodevelopmental disorders (NDDs) is chromatin modification and the genes that regulate chromatin. AT-rich interactive domain 1B (ARID1B), a chromatin modifier, has been linked to autism spectrum disorder and to affect rare and inherited genetic variation in a broad set of NDDs. METHODS A novel preclinical mouse model of Arid1b deficiency was created and validated to characterize and define neuroanatomical, behavioral and transcriptional phenotypes. Neuroanatomy was assessed ex vivo in adult animals and in vivo longitudinally from birth to adulthood. Behavioral testing was also performed throughout development and tested all aspects of motor, learning, sociability, repetitive behaviors, seizure susceptibility, and general milestones delays. RESULTS We validated decreased Arid1b mRNA and protein in Arid1b+/- mice, with signatures of increased axonal and synaptic gene expression, decreased transcriptional regulator and RNA processing expression in adult Arid1b+/- cerebellum. During neonatal development, Arid1b+/- mice exhibited robust impairments in ultrasonic vocalizations (USVs) and metrics of developmental growth. In addition, a striking sex effect was observed neuroanatomically throughout development. Behaviorally, as adults, Arid1b+/- mice showed low motor skills in open field exploration and normal three-chambered approach. Arid1b+/- mice had learning and memory deficits in novel object recognition but not in visual discrimination and reversal touchscreen tasks. Social interactions in the male-female social dyad with USVs revealed social deficits on some but not all parameters. No repetitive behaviors were observed. Brains of adult Arid1b+/- mice had a smaller cerebellum and a larger hippocampus and corpus callosum. The corpus callosum increase seen here contrasts previous reports which highlight losses in corpus callosum volume in mice and humans. LIMITATIONS The behavior and neuroimaging analyses were done on separate cohorts of mice, which did not allow a direct correlation between the imaging and behavioral findings, and the transcriptomic analysis was exploratory, with no validation of altered expression beyond Arid1b. CONCLUSIONS This study represents a full validation and investigation of a novel model of Arid1b+/- haploinsufficiency throughout development and highlights the importance of examining both sexes throughout development in NDDs.
Collapse
Affiliation(s)
- J Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, ON, M5T 3H7, Canada.
| | - S P Petkova
- Department of Psychiatry and Behavioral Sciences, MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Neuroscience Graduate Group, University of California - Davis, Davis, CA, USA
| | - A Kinman
- Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, ON, M5T 3H7, Canada
| | - L R Qiu
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neuroscience, The University of Oxford, Oxford, UK
| | - A Adhikari
- Department of Psychiatry and Behavioral Sciences, MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - A A Wade
- Neuroscience Graduate Group, University of California - Davis, Davis, CA, USA
| | - D Fernandes
- Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, ON, M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Z Lindenmaier
- Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, ON, M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - A Creighton
- The Centre for Phenogenomics, Hospital for Sick Children, Toronto, ON, Canada
| | - L M J Nutter
- The Centre for Phenogenomics, Hospital for Sick Children, Toronto, ON, Canada
| | - A S Nord
- Department of Psychiatry and Behavioral Sciences, MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Neuroscience Graduate Group, University of California - Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California - Davis, Davis, CA, USA
| | - J L Silverman
- Department of Psychiatry and Behavioral Sciences, MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - J P Lerch
- Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, ON, M5T 3H7, Canada
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neuroscience, The University of Oxford, Oxford, UK
| |
Collapse
|
92
|
Canales CP, Estes ML, Cichewicz K, Angara K, Aboubechara JP, Cameron S, Prendergast K, Su-Feher L, Zdilar I, Kreun EJ, Connolly EC, Seo JM, Goon JB, Farrelly K, Stradleigh TW, van der List D, Haapanen L, Van de Water J, Vogt D, McAllister AK, Nord AS. Sequential perturbations to mouse corticogenesis following in utero maternal immune activation. eLife 2021; 10:e60100. [PMID: 33666173 PMCID: PMC7979158 DOI: 10.7554/elife.60100] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 03/02/2021] [Indexed: 12/13/2022] Open
Abstract
In utero exposure to maternal immune activation (MIA) is an environmental risk factor for neurodevelopmental and neuropsychiatric disorders. Animal models provide an opportunity to identify mechanisms driving neuropathology associated with MIA. We performed time-course transcriptional profiling of mouse cortical development following induced MIA via poly(I:C) injection at E12.5. MIA-driven transcriptional changes were validated via protein analysis, and parallel perturbations to cortical neuroanatomy were identified via imaging. MIA-induced acute upregulation of genes associated with hypoxia, immune signaling, and angiogenesis, by 6 hr following exposure. This acute response was followed by changes in proliferation, neuronal and glial specification, and cortical lamination that emerged at E14.5 and peaked at E17.5. Decreased numbers of proliferative cells in germinal zones and alterations in neuronal and glial populations were identified in the MIA-exposed cortex. Overall, paired transcriptomic and neuroanatomical characterization revealed a sequence of perturbations to corticogenesis driven by mid-gestational MIA.
Collapse
Affiliation(s)
| | - Myka L Estes
- Center for Neuroscience, UC DavisDavisUnited States
| | | | - Kartik Angara
- Department of Pediatrics & Human Development, Michigan State UniversityEast LansingUnited States
| | | | | | | | | | - Iva Zdilar
- Center for Neuroscience, UC DavisDavisUnited States
| | | | | | | | - Jack B Goon
- Center for Neuroscience, UC DavisDavisUnited States
| | | | | | | | - Lori Haapanen
- Division of Rheumatology, Allergy and Clinical Immunology, UC DavisDavisUnited States
| | - Judy Van de Water
- Division of Rheumatology, Allergy and Clinical Immunology, UC DavisDavisUnited States
| | - Daniel Vogt
- Department of Pediatrics & Human Development, Michigan State UniversityEast LansingUnited States
| | | | - Alex S Nord
- Center for Neuroscience, UC DavisDavisUnited States
| |
Collapse
|
93
|
Mossink B, Negwer M, Schubert D, Nadif Kasri N. The emerging role of chromatin remodelers in neurodevelopmental disorders: a developmental perspective. Cell Mol Life Sci 2021; 78:2517-2563. [PMID: 33263776 PMCID: PMC8004494 DOI: 10.1007/s00018-020-03714-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/04/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022]
Abstract
Neurodevelopmental disorders (NDDs), including intellectual disability (ID) and autism spectrum disorders (ASD), are a large group of disorders in which early insults during brain development result in a wide and heterogeneous spectrum of clinical diagnoses. Mutations in genes coding for chromatin remodelers are overrepresented in NDD cohorts, pointing towards epigenetics as a convergent pathogenic pathway between these disorders. In this review we detail the role of NDD-associated chromatin remodelers during the developmental continuum of progenitor expansion, differentiation, cell-type specification, migration and maturation. We discuss how defects in chromatin remodelling during these early developmental time points compound over time and result in impaired brain circuit establishment. In particular, we focus on their role in the three largest cell populations: glutamatergic neurons, GABAergic neurons, and glia cells. An in-depth understanding of the spatiotemporal role of chromatin remodelers during neurodevelopment can contribute to the identification of molecular targets for treatment strategies.
Collapse
Affiliation(s)
- Britt Mossink
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Geert Grooteplein 10, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands
| | - Moritz Negwer
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Geert Grooteplein 10, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands
| | - Dirk Schubert
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Geert Grooteplein 10, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
94
|
Hurley S, Mohan C, Suetterlin P, Ellingford R, Riegman KLH, Ellegood J, Caruso A, Michetti C, Brock O, Evans R, Rudari F, Delogu A, Scattoni ML, Lerch JP, Fernandes C, Basson MA. Distinct, dosage-sensitive requirements for the autism-associated factor CHD8 during cortical development. Mol Autism 2021; 12:16. [PMID: 33627187 PMCID: PMC7905672 DOI: 10.1186/s13229-020-00409-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND CHD8 haploinsufficiency causes autism and macrocephaly with high penetrance in the human population. Chd8 heterozygous mice exhibit relatively subtle brain overgrowth and little gene expression changes in the embryonic neocortex. The purpose of this study was to generate new, sub-haploinsufficient Chd8 mouse models to allow us to identify and study the functions of CHD8 during embryonic cortical development. METHODS To examine the possibility that certain phenotypes may only appear at sub-heterozygous Chd8 levels in the mouse, we created an allelic series of Chd8-deficient mice to reduce CHD8 protein levels to approximately 35% (mild hypomorph), 10% (severe hypomorph) and 0% (neural-specific conditional knockout) of wildtype levels. We used RNA sequencing to compare transcriptional dysregulation, structural MRI and brain weight to investigate effects on brain size, and cell proliferation, differentiation and apoptosis markers in immunostaining assays to quantify changes in neural progenitor fate. RESULTS Mild Chd8 hypomorphs displayed significant postnatal lethality, with surviving animals exhibiting more pronounced brain hyperplasia than heterozygotes. Over 2000 genes were dysregulated in mild hypomorphs, including autism-associated neurodevelopmental and cell cycle genes. We identify increased proliferation of non-ventricular zone TBR2+ intermediate progenitors as one potential cause of brain hyperplasia in these mutants. Severe Chd8 hypomorphs displayed even greater transcriptional dysregulation, including evidence for p53 pathway upregulation. In contrast to mild hypomorphs, these mice displayed reduced brain size and increased apoptosis in the embryonic neocortex. Homozygous, conditional deletion of Chd8 in early neuronal progenitors resulted in pronounced brain hypoplasia, partly caused by p53 target gene derepression and apoptosis in the embryonic neocortex. Limitations Our findings identify an important role for the autism-associated factor CHD8 in controlling the proliferation of intermediate progenitors in the mouse neocortex. We propose that CHD8 has a similar function in human brain development, but studies on human cells are required to confirm this. Because many of our mouse mutants with reduced CHD8 function die shortly after birth, it is not possible to fully determine to what extent reduced CHD8 function results in autism-associated behaviours in mice. CONCLUSIONS Together, these findings identify important, dosage-sensitive functions for CHD8 in p53 pathway repression, neurodevelopmental gene expression and neural progenitor fate in the embryonic neocortex. We conclude that brain development is acutely sensitive to reduced CHD8 expression and that the varying sensitivities of different progenitor populations and cellular processes to CHD8 dosage result in non-linear effects on gene transcription and brain growth. Shaun Hurley, Conor Mohan and Philipp Suetterlin have contributed equally to this work.
Collapse
Affiliation(s)
- Shaun Hurley
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Conor Mohan
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Philipp Suetterlin
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Robert Ellingford
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | | | - Jacob Ellegood
- Department of Medical Biophysics, Mouse Imaging Centre, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Angela Caruso
- Department of Cell Biology and Neuroscience, Neurotoxicology and Neuroendocrinology Section, Istituto Superiore Di Sanità, Rome, Italy
- Department of Psychology, School of Behavioural Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Caterina Michetti
- Department of Cell Biology and Neuroscience, Neurotoxicology and Neuroendocrinology Section, Istituto Superiore Di Sanità, Rome, Italy
- Centre for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Genova, Italy
| | - Olivier Brock
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Romy Evans
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Fabrizio Rudari
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Alessio Delogu
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Maria Luisa Scattoni
- Department of Cell Biology and Neuroscience, Neurotoxicology and Neuroendocrinology Section, Istituto Superiore Di Sanità, Rome, Italy
| | - Jason P Lerch
- Department of Medical Biophysics, Mouse Imaging Centre, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Cathy Fernandes
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - M Albert Basson
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
95
|
Kweon H, Jung WB, Im GH, Ryoo J, Lee JH, Do H, Choi Y, Song YH, Jung H, Park H, Qiu LR, Ellegood J, Shim HJ, Yang E, Kim H, Lerch JP, Lee SH, Chung WS, Kim D, Kim SG, Kim E. Excitatory neuronal CHD8 in the regulation of neocortical development and sensory-motor behaviors. Cell Rep 2021; 34:108780. [PMID: 33626347 DOI: 10.1016/j.celrep.2021.108780] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/16/2020] [Accepted: 02/02/2021] [Indexed: 12/31/2022] Open
Abstract
CHD8 (chromodomain helicase DNA-binding protein 8) is a chromatin remodeler associated with autism spectrum disorders. Homozygous Chd8 deletion in mice leads to embryonic lethality, making it difficult to assess whether CHD8 regulates brain development and whether CHD8 haploinsufficiency-related macrocephaly reflects normal CHD8 functions. Here, we report that homozygous conditional knockout of Chd8 restricted to neocortical glutamatergic neurons causes apoptosis-dependent near-complete elimination of neocortical structures. These mice, however, display normal survival and hyperactivity, anxiolytic-like behavior, and increased social interaction. They also show largely normal auditory function and moderately impaired visual and motor functions but enhanced whisker-related somatosensory function. These changes accompany thalamic hyperactivity, revealed by 15.2-Tesla fMRI, and increased intrinsic excitability and decreased inhibitory synaptic transmission in thalamic ventral posterior medial (VPM) neurons involved in somatosensation. These results suggest that excitatory neuronal CHD8 critically regulates neocortical development through anti-apoptotic mechanisms, neocortical elimination distinctly affects cognitive behaviors and sensory-motor functions in mice, and Chd8 haploinsufficiency-related macrocephaly might represent compensatory responses.
Collapse
Affiliation(s)
- Hanseul Kweon
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Won Beom Jung
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Geun Ho Im
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea
| | - Jia Ryoo
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Joon-Hyuk Lee
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Hogyeong Do
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - Yeonsoo Choi
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - You-Hyang Song
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Hwajin Jung
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - Haram Park
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - Lily R Qiu
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON M5T 3H7, Canada; Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford OX3 9DU, UK
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON M5T 3H7, Canada; Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford OX3 9DU, UK
| | - Hyun-Ji Shim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea
| | - Esther Yang
- Department of Anatomy and Division of Brain Korea 21, Biomedical Science, College of Medicine, Korea University, Seoul 02841, Korea
| | - Hyun Kim
- Department of Anatomy and Division of Brain Korea 21, Biomedical Science, College of Medicine, Korea University, Seoul 02841, Korea
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON M5T 3H7, Canada; Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford OX3 9DU, UK
| | - Seung-Hee Lee
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea.
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea.
| |
Collapse
|
96
|
Copping NA, Silverman JL. Abnormal electrophysiological phenotypes and sleep deficits in a mouse model of Angelman Syndrome. Mol Autism 2021; 12:9. [PMID: 33549123 PMCID: PMC7866697 DOI: 10.1186/s13229-021-00416-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/18/2021] [Indexed: 01/17/2023] Open
Abstract
Background Angelman Syndrome (AS) is a rare genetic disorder characterized by impaired communication, motor and balance deficits, intellectual disabilities, recurring seizures and abnormal sleep patterns. The genetic cause of AS is neuronal-specific loss of expression of UBE3A (ubiquitin-protein ligase E6-AP), an imprinted gene. Seizure and sleep disorders are highly prevalent (> 80%) in the AS population. The present experiments were designed to identify translational, neurophysiological outcome measures in a model of AS. Methods We used the exon-2 deletion mouse (Ube3a-del) on a C57BL/6J background to assess seizure, sleep and electrophysiological phenotypes. Seizure susceptibility has been reported in Ube3a-del mice with a variety of seizure induction methods. Here, we provoked seizures by a single high-dose injection of 80 mg/kg pentylenetetrazole. Novel experiments included the utilization of wireless telemetry devices to acquire global electroencephalogram (EEG) and neurophysiological data on electrographic seizures, power spectra, light–dark cycles, sleep stages and sleep spindles in Ube3a-del and WT mice. Results Ube3a-del mice exhibited reduced seizure threshold compared to WT. EEG illustrated that Ube3a-del mice had increased epileptiform spiking activity and delta power, which corroborates findings from other laboratories and recapitulates clinical reports in AS. This is the first report to use a cortical surface-based recording by a wireless telemetry device over tethered/fixed head-mount depth recordings. Less time in both paradoxical and slow-wave sleep, longer latencies to paradoxical sleep stages and total less sleep time in Ube3a-del mice were observed compared to WT. For the first time, we detected fewer sleep spindles in the AS mouse model. Limitations This study was limited to the exon 2 deletion mouse model, and future work will investigate the rat model of AS, containing a complete Ube3a deletion and pair EEG with behavior. Conclusions Our data enhance rigor and translatability as our study provides important corroboration of previous reports on epileptiform and elevated delta power. For the first time we report neurophysiological phenotypes collected via translational methodology. Furthermore, this is the first report of reduced sleep spindles, a critical marker of memory consolidation during sleep, in an AS model. Our results are useful outcomes for therapeutic testing.
Collapse
Affiliation(s)
- N A Copping
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001B, Research II Building 96, 4625 2nd Avenue, Sacramento, CA, 95817, USA
| | - J L Silverman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001B, Research II Building 96, 4625 2nd Avenue, Sacramento, CA, 95817, USA.
| |
Collapse
|
97
|
Dhindsa RS, Zoghbi AW, Krizay DK, Vasavda C, Goldstein DB. A Transcriptome-Based Drug Discovery Paradigm for Neurodevelopmental Disorders. Ann Neurol 2021; 89:199-211. [PMID: 33159466 PMCID: PMC8122510 DOI: 10.1002/ana.25950] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022]
Abstract
Advances in genetic discoveries have created substantial opportunities for precision medicine in neurodevelopmental disorders. Many of the genes implicated in these diseases encode proteins that regulate gene expression, such as chromatin-associated proteins, transcription factors, and RNA-binding proteins. The identification of targeted therapeutics for individuals carrying mutations in these genes remains a challenge, as the encoded proteins can theoretically regulate thousands of downstream targets in a considerable number of cell types. Here, we propose the application of a drug discovery approach originally developed for cancer called "transcriptome reversal" for these neurodevelopmental disorders. This approach attempts to identify compounds that reverse gene-expression signatures associated with disease states. ANN NEUROL 2021;89:199-211.
Collapse
Affiliation(s)
- Ryan S. Dhindsa
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, USA
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Anthony W. Zoghbi
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, USA; New York State Psychiatric Institute, New York, USA
| | - Daniel K. Krizay
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, USA
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, USA
| | - Chirag Vasavda
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David B. Goldstein
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, USA
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, USA
| |
Collapse
|
98
|
Hoffmann A, Spengler D. Chromatin Remodeler CHD8 in Autism and Brain Development. J Clin Med 2021; 10:366. [PMID: 33477995 PMCID: PMC7835889 DOI: 10.3390/jcm10020366] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/14/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022] Open
Abstract
Chromodomain Helicase DNA-binding 8 (CHD8) is a high confidence risk factor for autism spectrum disorders (ASDs) and the genetic cause of a distinct neurodevelopmental syndrome with the core symptoms of autism, macrocephaly, and facial dysmorphism. The role of CHD8 is well-characterized at the structural, biochemical, and transcriptional level. By contrast, much less is understood regarding how mutations in CHD8 underpin altered brain function and mental disease. Studies on various model organisms have been proven critical to tackle this challenge. Here, we scrutinize recent advances in this field with a focus on phenotypes in transgenic animal models and highlight key findings on neurodevelopment, neuronal connectivity, neurotransmission, synaptic and homeostatic plasticity, and habituation. Against this backdrop, we further discuss how to improve future animal studies, both in terms of technical issues and with respect to the sex-specific effects of Chd8 mutations for neuronal and higher-systems level function. We also consider outstanding questions in the field including 'humanized' mice models, therapeutic interventions, and how the use of pluripotent stem cell-derived cerebral organoids might help to address differences in neurodevelopment trajectories between model organisms and humans.
Collapse
Affiliation(s)
| | - Dietmar Spengler
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, 80804 Munich, Germany;
| |
Collapse
|
99
|
Morphological study of embryonic Chd8 +/- mouse brains using light-sheet microscopy. BMC Res Notes 2021; 14:23. [PMID: 33436073 PMCID: PMC7802325 DOI: 10.1186/s13104-020-05436-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/28/2020] [Indexed: 01/24/2023] Open
Abstract
Objective Autism spectrum disorder (ASD) encompasses a group of neurodevelopmental conditions that remain poorly understood due to their genetic complexity. CHD8 is a risk allele strongly associated with ASD, and heterozygous Chd8 loss-of-function mice have been reported to exhibit macrocephaly in early postnatal stages. In this work, we sought to identify measurable brain alterations in early embryonic development. Results We performed light-sheet fluorescence microscopy imaging of N-cadherin stained and optically cleared Chd8+/− and wild-type mouse brains at embryonic day 12.5 (E12.5). We report a detailed morphometric characterization of embryonic brain shapes and cortical neuroepithelial apical architecture. While Chd8+/− characteristic expansion of the forebrain and midbrain was not observed this early in embryogenesis, a tendency for a decreased lateral ventricular sphericity and an increased intraocular distance in Chd8+/− brains was found compared to controls. This study advocates the use of high-resolution microscopy technologies and multi-scale morphometric analyses of target brain regions to explore the etiology and cellular basis of Chd8 haploinsufficiency.
Collapse
|
100
|
Alonso-Gonzalez A, Calaza M, Amigo J, González-Peñas J, Martínez-Regueiro R, Fernández-Prieto M, Parellada M, Arango C, Rodriguez-Fontenla C, Carracedo A. Exploring the biological role of postzygotic and germinal de novo mutations in ASD. Sci Rep 2021; 11:319. [PMID: 33431980 PMCID: PMC7801448 DOI: 10.1038/s41598-020-79412-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
De novo mutations (DNMs), including germinal and postzygotic mutations (PZMs), are a strong source of causality for Autism Spectrum Disorder (ASD). However, the biological processes involved behind them remain unexplored. Our aim was to detect DNMs (germinal and PZMs) in a Spanish ASD cohort (360 trios) and to explore their role across different biological hierarchies (gene, biological pathway, cell and brain areas) using bioinformatic approaches. For the majority of the analysis, a combined ASD cohort (N = 2171 trios) was created using previously published data by the Autism Sequencing Consortium (ASC). New plausible candidate genes for ASD such as FMR1 and NFIA were found. In addition, genes harboring PZMs were significantly enriched for miR-137 targets in comparison with germinal DNMs that were enriched in GO terms related to synaptic transmission. The expression pattern of genes with PZMs was restricted to early mid-fetal cortex. In contrast, the analysis of genes with germinal DNMs revealed a spatio-temporal window from early to mid-fetal development stages, with expression in the amygdala, cerebellum, cortex and striatum. These results provide evidence of the pathogenic role of PZMs and suggest the existence of distinct mechanisms between PZMs and germinal DNMs that are influencing ASD risk.
Collapse
Affiliation(s)
- A Alonso-Gonzalez
- Grupo de Medicina Xenómica, Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (FIDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Genomics and Bioinformatics Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Av Barcelona 31, 15706, Santiago de Compostela, Spain
| | - M Calaza
- Grupo de Medicina Xenómica, Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (FIDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Genomics and Bioinformatics Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Av Barcelona 31, 15706, Santiago de Compostela, Spain
| | - J Amigo
- Fundación Pública Galega de Medicina Xenómica (FPGMX), Centro de Investigación Biomédica en Red, Enfermedades Raras (CIBERER), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - J González-Peñas
- Centro De Investigación Biomédica en Red de Salud Mental (CIBERSAM), Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, IiSGM, School of Medicine, Universidad Complutense, Madrid, Spain
| | - R Martínez-Regueiro
- Grupo de Medicina Xenómica, Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (FIDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Genomics and Bioinformatics Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Av Barcelona 31, 15706, Santiago de Compostela, Spain
| | - M Fernández-Prieto
- Grupo de Medicina Xenómica, Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (FIDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Genomics and Bioinformatics Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Av Barcelona 31, 15706, Santiago de Compostela, Spain
| | - M Parellada
- Centro De Investigación Biomédica en Red de Salud Mental (CIBERSAM), Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, IiSGM, School of Medicine, Universidad Complutense, Madrid, Spain
| | - C Arango
- Centro De Investigación Biomédica en Red de Salud Mental (CIBERSAM), Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, IiSGM, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Cristina Rodriguez-Fontenla
- Genomics and Bioinformatics Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Av Barcelona 31, 15706, Santiago de Compostela, Spain.
| | - A Carracedo
- Grupo de Medicina Xenómica, Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (FIDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Genomics and Bioinformatics Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Av Barcelona 31, 15706, Santiago de Compostela, Spain.,Fundación Pública Galega de Medicina Xenómica (FPGMX), Centro de Investigación Biomédica en Red, Enfermedades Raras (CIBERER), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|