Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Mol Neurobiol ; 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38652351

ABSTRACT

Neuronal hyperexcitability within developing cortical circuits is a common characteristic of several heritable neurodevelopmental disorders, including Fragile X Syndrome (FXS), intellectual disability and autism spectrum disorders (ASD). While this aberrant circuitry is typically studied from a neuron-centric perspective, glial cells secrete soluble factors that regulate both neurite extension and synaptogenesis during development. The nucleotide-mediated purinergic signalling system is particularly instrumental in facilitating these effects. We recently reported that within a FXS animal model, the Fmr1 KO mouse, the purinergic signalling system is upregulated in cortical astrocytes leading to altered secretion of synaptogenic and plasticity-related proteins. In this study, we examined whether elevated astrocyte purinergic signalling also impacts neuronal morphology and connectivity of Fmr1 KO cortical neurons. Here, we found that conditioned media from primary Fmr1 KO astrocytes was sufficient to enhance neurite extension and complexity of both wildtype and Fmr1 KO neurons to a similar degree as UTP-mediated outgrowth. Significantly enhanced firing was also observed in Fmr1 KO neuron-astrocyte co-cultures grown on microelectrode arrays but was associated with large deficits in firing synchrony. The selective P2Y2 purinergic receptor antagonist AR-C 118925XX effectively normalized much of the aberrant Fmr1 KO activity, designating P2Y2 as a potential therapeutic target in FXS. These results not only demonstrate the importance of astrocyte soluble factors in the development of neural circuitry, but also show that P2Y purinergic receptors play a distinct role in pathological FXS neuronal activity.

2.
Sci Adv ; 10(15): eadf7001, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38608030

ABSTRACT

Genes implicated in translation control have been associated with autism spectrum disorders (ASDs). However, some important genetic causes of autism, including the 16p11.2 microdeletion, bear no obvious connection to translation. Here, we use proteomics, genetics, and translation assays in cultured cells and mouse brain to reveal altered translation mediated by loss of the kinase TAOK2 in 16p11.2 deletion models. We show that TAOK2 associates with the translational machinery and functions as a translational brake by phosphorylating eukaryotic elongation factor 2 (eEF2). Previously, all signal-mediated regulation of translation elongation via eEF2 phosphorylation was believed to be mediated by a single kinase, eEF2K. However, we show that TAOK2 can directly phosphorylate eEF2 on the same regulatory site, but functions independently of eEF2K signaling. Collectively, our results reveal an eEF2K-independent signaling pathway for control of translation elongation and suggest altered translation as a molecular component in the etiology of some forms of ASD.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Ursidae , Animals , Mice , Autistic Disorder/genetics , Peptide Elongation Factor 2 , Phosphorylation , Autism Spectrum Disorder/genetics , Biological Assay
3.
Transl Psychiatry ; 13(1): 217, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37344450

ABSTRACT

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder caused by genetic or environmental perturbations during early development. Diagnoses are dependent on the identification of behavioral abnormalities that likely emerge well after the disorder is established, leaving critical developmental windows uncharacterized. This is further complicated by the incredible clinical and genetic heterogeneity of the disorder that is not captured in most mammalian models. In recent years, advancements in stem cell technology have created the opportunity to model ASD in a human context through the use of pluripotent stem cells (hPSCs), which can be used to generate 2D cellular models as well as 3D unguided- and region-specific neural organoids. These models produce profoundly intricate systems, capable of modeling the developing brain spatiotemporally to reproduce key developmental milestones throughout early development. When complemented with multi-omics, genome editing, and electrophysiology analysis, they can be used as a powerful tool to profile the neurobiological mechanisms underlying this complex disorder. In this review, we will explore the recent advancements in hPSC-based modeling, discuss present and future applications of the model to ASD research, and finally consider the limitations and future directions within the field to make this system more robust and broadly applicable.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Animals , Humans , Autistic Disorder/genetics , Autism Spectrum Disorder/genetics , Induced Pluripotent Stem Cells/physiology , Organoids , Mammals
4.
Mol Psychiatry ; 28(4): 1747-1769, 2023 04.
Article in English | MEDLINE | ID: mdl-36604605

ABSTRACT

Copy number variations (CNVs) are associated with psychiatric and neurodevelopmental disorders (NDDs), and most, including the recurrent 15q13.3 microdeletion disorder, have unknown disease mechanisms. We used a heterozygous 15q13.3 microdeletion mouse model and patient iPSC-derived neurons to reveal developmental defects in neuronal maturation and network activity. To identify the underlying molecular dysfunction, we developed a neuron-specific proximity-labeling proteomics (BioID2) pipeline, combined with patient mutations, to target the 15q13.3 CNV genetic driver OTUD7A. OTUD7A is an emerging independent NDD risk gene with no known function in the brain, but has putative deubiquitinase function. The OTUD7A protein-protein interaction network included synaptic, axonal, and cytoskeletal proteins and was enriched for ASD and epilepsy risk genes (Ank3, Ank2, SPTAN1, SPTBN1). The interactions between OTUD7A and Ankyrin-G (Ank3) and Ankyrin-B (Ank2) were disrupted by an epilepsy-associated OTUD7A L233F variant. Further investigation of Ankyrin-G in mouse and human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed protein instability, increased polyubiquitination, and decreased levels in the axon initial segment, while structured illumination microscopy identified reduced Ankyrin-G nanodomains in dendritic spines. Functional analysis of human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed shared and distinct impairments to axonal growth and intrinsic excitability. Importantly, restoring OTUD7A or Ankyrin-G expression in 15q13.3 microdeletion neurons led to a reversal of abnormalities. These data reveal a critical OTUD7A-Ankyrin pathway in neuronal development, which is impaired in the 15q13.3 microdeletion syndrome, leading to neuronal dysfunction. Furthermore, our study highlights the utility of targeting CNV genes using cell type-specific proteomics to identify shared and unexplored disease mechanisms across NDDs.


Subject(s)
Ankyrins , Epilepsy , Humans , Mice , Animals , Ankyrins/genetics , DNA Copy Number Variations , Epilepsy/genetics , Neurons
5.
Front Cell Neurosci ; 17: 1239069, 2023.
Article in English | MEDLINE | ID: mdl-38293651

ABSTRACT

SCN2A is an autism spectrum disorder (ASD) risk gene and encodes a voltage-gated sodium channel. However, the impact of ASD-associated SCN2A de novo variants on human neuron development is unknown. We studied SCN2A using isogenic SCN2A-/- induced pluripotent stem cells (iPSCs), and patient-derived iPSCs harboring a de novo R607* truncating variant. We used Neurogenin2 to generate excitatory (glutamatergic) neurons and found that SCN2A+/R607* and SCN2A-/- neurons displayed a reduction in synapse formation and excitatory synaptic activity. We found differential impact on actional potential dynamics and neuronal excitability that reveals a loss-of-function effect of the R607* variant. Our study reveals that a de novo truncating SCN2A variant impairs the development of human neuronal function.

6.
Cell Rep ; 41(8): 111678, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36417873

ABSTRACT

There are hundreds of risk genes associated with autism spectrum disorder (ASD), but signaling networks at the protein level remain unexplored. We use neuron-specific proximity-labeling proteomics (BioID2) to identify protein-protein interaction (PPI) networks for 41 ASD risk genes. Neuron-specific PPI networks, including synaptic transmission proteins, are disrupted by de novo missense variants. The PPI network map reveals convergent pathways, including mitochondrial/metabolic processes, Wnt signaling, and MAPK signaling. CRISPR knockout displays an association between mitochondrial activity and ASD risk genes. The PPI network shows an enrichment of 112 additional ASD risk genes and differentially expressed genes from postmortem ASD patients. Clustering of risk genes based on PPI networks identifies gene groups corresponding to clinical behavior score severity. Our data report that cell type-specific PPI networks can identify individual and convergent ASD signaling networks, provide a method to assess patient variants, and highlight biological insight into disease mechanisms and sub-cohorts of ASD.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Humans , Autistic Disorder/genetics , Autism Spectrum Disorder/genetics , Protein Interaction Maps/genetics , Neurons , Proteins , Wnt Signaling Pathway
7.
Mol Psychiatry ; 27(11): 4707-4721, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36123424

ABSTRACT

The precise development of the neocortex is a prerequisite for higher cognitive and associative functions. Despite numerous advances that have been made in understanding neuronal differentiation and cortex development, our knowledge regarding the impact of specific genes associated with neurodevelopmental disorders on these processes is still limited. Here, we show that Taok2, which is encoded in humans within the autism spectrum disorder (ASD) susceptibility locus 16p11.2, is essential for neuronal migration. Overexpression of de novo mutations or rare variants from ASD patients disrupts neuronal migration in an isoform-specific manner. The mutated TAOK2α variants but not the TAOK2ß variants impaired neuronal migration. Moreover, the TAOK2α isoform colocalizes with microtubules. Consequently, neurons lacking Taok2 have unstable microtubules with reduced levels of acetylated tubulin and phosphorylated JNK1. Mice lacking Taok2 develop gross cortical and cortex layering abnormalities. Moreover, acute Taok2 downregulation or Taok2 knockout delayed the migration of upper-layer cortical neurons in mice, and the expression of a constitutively active form of JNK1 rescued these neuronal migration defects. Finally, we report that the brains of the Taok2 KO and 16p11.2 del Het mouse models show striking anatomical similarities and that the heterozygous 16p11.2 microdeletion mouse model displayed reduced levels of phosphorylated JNK1 and neuronal migration deficits, which were ameliorated upon the introduction of TAOK2α in cortical neurons and in the developing cortex of those mice. These results delineate the critical role of TAOK2 in cortical development and its contribution to neurodevelopmental disorders, including ASD.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Neocortex , Protein Serine-Threonine Kinases , Animals , Humans , Mice , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Disease Models, Animal , Microtubules/genetics , Microtubules/metabolism , Neocortex/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
8.
J Mol Evol ; 89(4-5): 195-213, 2021 06.
Article in English | MEDLINE | ID: mdl-33630117

ABSTRACT

Sexual dimorphism or sex bias in diseases and mental disorders have two biological causes: sexual selection and sex hormones. We review the role of sexual selection theory and bring together decades of molecular studies on the variation and evolution of sex-biased genes and provide a theoretical basis for the causes of sex bias in disease and health. We present a Sexual Selection-Sex Hormone theory and show that male-driven evolution, including sexual selection, leads to: (1) increased male vulnerability due to negative pleiotropic effects associated with male-driven sexual selection and evolution; (2) increased rates of male-driven mutations and epimutations in response to early fitness gains and at the cost of late fitness; and (3) enhanced female immunity due to antagonistic responses to mutations that are beneficial to males but harmful to females, reducing female vulnerability to diseases and increasing the thresholds for disorders such as autism. Female-driven evolution, such as reproduction-related fluctuation in female sex hormones in association with stress and social condition, has been shown to be associated with increased risk of certain mental disorders such as major depression disorder in women. Bodies have history, cells have memories. An evolutionary framework, such as the Sexual Selection-Sex Hormone theory, provides a historical perspective for understanding how the differences in the sex-biased diseases and mental disorders have evolved over time. It has the potential to direct the development of novel preventive and treatment strategies.


Subject(s)
Mental Disorders , Sexism , Female , Humans , Male , Mental Disorders/genetics , Reproduction , Selection, Genetic , Sex Characteristics
9.
J Proteome Res ; 20(1): 1052-1062, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33337894

ABSTRACT

DIX-domain containing 1 (Dixdc1) is an important regulator of neuronal development including cortical neurogenesis, neuronal migration and synaptic connectivity, and sequence variants in the gene have been linked to autism spectrum disorders (ASDs). Previous studies indicate that Dixdc1 controls neurogenesis through Wnt signaling, whereas its regulation of dendrite and synapse development requires Wnt and cytoskeletal signaling. However, the prediction of these signaling pathways is primarily based on the structure of Dixdc1. Given the role of Dixdc1 in neural development and brain disorders, we hypothesized that Dixdc1 may regulate additional signaling pathways in the brain. We performed transcriptomic and proteomic analyses of Dixdc1 KO mouse cortices to reveal such alterations. We found that transcriptomic approaches do not yield any novel findings about the downstream impacts of Dixdc1. In comparison, our proteomic approach reveals that several important mitochondrial proteins are significantly dysregulated in the absence of Dixdc1, suggesting a novel function of Dixdc1.


Subject(s)
Autistic Disorder , Intracellular Signaling Peptides and Proteins , Animals , Cell Movement , Mice , Microfilament Proteins , Proteomics
10.
Nat Neurosci ; 23(9): 1102-1110, 2020 09.
Article in English | MEDLINE | ID: mdl-32661395

ABSTRACT

Cerebellar dysfunction has been demonstrated in autism spectrum disorders (ASDs); however, the circuits underlying cerebellar contributions to ASD-relevant behaviors remain unknown. In this study, we demonstrated functional connectivity between the cerebellum and the medial prefrontal cortex (mPFC) in mice; showed that the mPFC mediates cerebellum-regulated social and repetitive/inflexible behaviors; and showed disruptions in connectivity between these regions in multiple mouse models of ASD-linked genes and in individuals with ASD. We delineated a circuit from cerebellar cortical areas Right crus 1 (Rcrus1) and posterior vermis through the cerebellar nuclei and ventromedial thalamus and culminating in the mPFC. Modulation of this circuit induced social deficits and repetitive behaviors, whereas activation of Purkinje cells (PCs) in Rcrus1 and posterior vermis improved social preference impairments and repetitive/inflexible behaviors, respectively, in male PC-Tsc1 mutant mice. These data raise the possibility that these circuits might provide neuromodulatory targets for the treatment of ASD.


Subject(s)
Autism Spectrum Disorder/physiopathology , Cerebellum/physiopathology , Neural Pathways/physiopathology , Prefrontal Cortex/physiopathology , Animals , Male , Mice , Mice, Mutant Strains
11.
Mol Autism ; 11(1): 27, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32317014

ABSTRACT

Proteomics is the large-scale study of the total protein content and their overall function within a cell through multiple facets of research. Advancements in proteomic methods have moved past the simple quantification of proteins to the identification of post-translational modifications (PTMs) and the ability to probe interactions between these proteins, spatially and temporally. Increased sensitivity and resolution of mass spectrometers and sample preparation protocols have drastically reduced the large amount of cells required and the experimental variability that had previously hindered its use in studying human neurological disorders. Proteomics offers a new perspective to study the altered molecular pathways and networks that are associated with autism spectrum disorders (ASD). The differences between the transcriptome and proteome, combined with the various types of post-translation modifications that regulate protein function and localization, highlight a novel level of research that has not been appropriately investigated. In this review, we will discuss strategies using proteomics to study ASD and other neurological disorders, with a focus on how these approaches can be combined with induced pluripotent stem cell (iPSC) studies. Proteomic analysis of iPSC-derived neurons have already been used to measure changes in the proteome caused by patient mutations, analyze changes in PTMs that resulted in altered biological pathways, and identify potential biomarkers. Further advancements in both proteomic techniques and human iPSC differentiation protocols will continue to push the field towards better understanding ASD disease pathophysiology. Proteomics using iPSC-derived neurons from individuals with ASD offers a window for observing the altered proteome, which is necessary in the future development of therapeutics against specific targets.


Subject(s)
Neurodegenerative Diseases/metabolism , Neurodevelopmental Disorders/metabolism , Proteomics/methods , Animals , Biomarkers/metabolism , Brain/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Protein Interaction Maps , Protein Processing, Post-Translational
12.
Clin Genet ; 97(4): 567-575, 2020 04.
Article in English | MEDLINE | ID: mdl-31997314

ABSTRACT

Heterozygous microdeletions of chromosome 15q13.3 (MIM: 612001) show incomplete penetrance and are associated with a highly variable phenotype that may include intellectual disability, epilepsy, facial dysmorphism and digit anomalies. Rare patients carrying homozygous deletions show more severe phenotypes including epileptic encephalopathy, hypotonia and poor growth. For years, CHRNA7 (MIM: 118511), was considered the candidate gene that could account for this syndrome. However, recent studies in mouse models have shown that OTUD7A/CEZANNE2 (MIM: 612024), which encodes for an ovarian tumor (OTU) deubiquitinase, should be considered the critical gene responsible for brain dysfunction. In this study, a patient presenting with severe global developmental delay, language impairment and epileptic encephalopathy was referred to our genetics center. Trio exome sequencing (tES) analysis identified a homozygous OTUD7A missense variant (NM_130901.2:c.697C>T), predicted to alter an ultraconserved amino acid, p.(Leu233Phe), lying within the OTU catalytic domain. Its subsequent segregation analysis revealed that the parents, presenting with learning disability, and brother were heterozygous carriers. Biochemical assays demonstrated that proteasome complex formation and function were significantly reduced in patient-derived fibroblasts and in OTUD7A knockout HAP1 cell line. We provide evidence that biallelic pathogenic OTUD7A variation is linked to early-onset epileptic encephalopathy and proteasome dysfunction.


Subject(s)
Chromosome Disorders/genetics , Deubiquitinating Enzymes/genetics , Epilepsy/genetics , Intellectual Disability/genetics , Seizures/genetics , Animals , Chromosome Deletion , Chromosome Disorders/physiopathology , Chromosomes, Human, Pair 15/genetics , Epilepsy/physiopathology , Female , Heterozygote , Homozygote , Humans , Intellectual Disability/pathology , Intellectual Disability/physiopathology , Male , Mice , Mutation, Missense/genetics , Phenotype , Seizures/physiopathology , Exome Sequencing , alpha7 Nicotinic Acetylcholine Receptor/genetics
13.
Can J Pain ; 4(4): 37-44, 2020 Dec 30.
Article in English | MEDLINE | ID: mdl-33987518

ABSTRACT

BACKGROUND: Pain is a complex neurobiological response with a multitude of causes; however, patients with autism spectrum disorder (ASD) often report chronic pain with no known etiology. Recent research has been aimed toward identifying the causal mechanisms of pain in mouse and human models of ASD. In recent years, efforts have been made to better document and explore secondary phenotypes observed in ASD patients in the clinic. As new sequencing studies have become more powered with larger cohorts within ASD, specific genes and their variants are often left uncharacterized or validated. In this review we highlight ASD risk genes often presented with pain comorbidities. AIMS: This mini-review bridges the gap between two fields of literature, neurodevelopmental disorders and pain research. We discuss the importance of the genetic landscape of ASD and its links to pain phenotypes. RESULTS: Among the numerous genes implicated in ASD, few have been implicated with varying severities of pain comorbidity. Mutations in these genes, such as SCN9A, SHANK3, and CNTNAP2, lead to altered neuronal function that produce different responses to pain, shown in both mouse and human models. CONCLUSION: There is a necessity to use new technologies to advance the current understanding of ASD risk genes and their contributions to pain. Secondly, there is a need to power future ASD risk genes associated with pain with their own cohort, because a better understanding is needed of this subpopulation.


Contexte: La douleur est une réponse neurobiologique complexe dont les causes sont multiples ; cependant, les patients atteints de troubles du spectre de l'autisme (TSA) rapportent souvent une douleur chronique sans étiologie connue. Des recherches récentes ont visé à identifier les mécanismes causaux de la douleur chez des modèles murins et humains.Ces dernières années, des efforts ont été faits pour mieux documenter et étudier les phénotypes secondaires observés chez les patients atteints de TSA en clinique. Étant donné que les nouvelles études de séquençage sont devenues plus puissantes et se réalisent avec des cohortes plus importantes au sein des TSA, des gènes spécifiques et leurs variantes demeurent souvent non caractérisés ou validés. Dans cette revue, nous mettons en évidence les gènes de risque de TSA qui se présentent souvent avec des comorbidités douloureuses.Objectifs: Cette mini-revue comble le fossé entre deux domaines de la littérature, les troubles neurodéveloppementaux et la recherche sur la douleur. Nous discutons de l'importance du paysage génétique des TSA et de ses liens avec les phénotypes de la douleur.Résultats: Parmi les nombreux gènes impliqués dans les TSA, peu ont été impliqués avec divers de degrés de sévérité de la comorbidité de la douleur. Des mutations dans ces gènes, tels que SCN9A, SHANK3 et CNTNAP2, conduisent à une fonction neuronale altérée qui produit des réponses différentes à la douleur, que l'on retrouve à la fois chez les modèles murins et humains.Conclusion: Il est nécessaire d'utiliser les nouvelles technologies pour faire progresser la compréhension actuelle des gènes de risque de TSA et leurs contributions à la douleur. Deuxièmement, il est nécessaire d'augmenter la puissance des futurs gènes de risque de TSA associés à la douleur avec leur propre cohorte, car une meilleure compréhension de cette sous-population est nécessaire.

14.
Elife ; 82019 02 12.
Article in English | MEDLINE | ID: mdl-30747104

ABSTRACT

Induced pluripotent stem cell (iPSC)-derived neurons are increasingly used to model Autism Spectrum Disorder (ASD), which is clinically and genetically heterogeneous. To study the complex relationship of penetrant and weaker polygenic risk variants to ASD, 'isogenic' iPSC-derived neurons are critical. We developed a set of procedures to control for heterogeneity in reprogramming and differentiation, and generated 53 different iPSC-derived glutamatergic neuronal lines from 25 participants from 12 unrelated families with ASD. Heterozygous de novo and rare-inherited presumed-damaging variants were characterized in ASD risk genes/loci. Combinations of putative etiologic variants (GLI3/KIF21A or EHMT2/UBE2I) in separate families were modeled. We used a multi-electrode array, with patch-clamp recordings, to determine a reproducible synaptic phenotype in 25% of the individuals with ASD (other relevant data on the remaining lines was collected). Our most compelling new results revealed a consistent spontaneous network hyperactivity in neurons deficient for CNTN5 or EHMT2. The biobank of iPSC-derived neurons and accompanying genomic data are available to accelerate ASD research. Editorial note: This article has been through an editorial process in which authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Subject(s)
Autistic Disorder/physiopathology , Contactins/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Induced Pluripotent Stem Cells/physiology , Nerve Net/physiology , Neurons/physiology , Adolescent , Adult , Cells, Cultured , Child , Contactins/deficiency , Contactins/genetics , Electrophysiological Phenomena , Female , Heterozygote , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Humans , Kinesins/genetics , Kinesins/metabolism , Male , Middle Aged , Models, Theoretical , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Young Adult , Zinc Finger Protein Gli3/genetics , Zinc Finger Protein Gli3/metabolism
16.
Mol Psychiatry ; 24(9): 1329-1350, 2019 09.
Article in English | MEDLINE | ID: mdl-29467497

ABSTRACT

Atypical brain connectivity is a major contributor to the pathophysiology of neurodevelopmental disorders (NDDs) including autism spectrum disorders (ASDs). TAOK2 is one of several genes in the 16p11.2 microdeletion region, but whether it contributes to NDDs is unknown. We performed behavioral analysis on Taok2 heterozygous (Het) and knockout (KO) mice and found gene dosage-dependent impairments in cognition, anxiety, and social interaction. Taok2 Het and KO mice also have dosage-dependent abnormalities in brain size and neural connectivity in multiple regions, deficits in cortical layering, dendrite and synapse formation, and reduced excitatory neurotransmission. Whole-genome and -exome sequencing of ASD families identified three de novo mutations in TAOK2 and functional analysis in mice and human cells revealed that all the mutations impair protein stability, but they differentially impact kinase activity, dendrite growth, and spine/synapse development. Mechanistically, loss of Taok2 activity causes a reduction in RhoA activation, and pharmacological enhancement of RhoA activity rescues synaptic phenotypes. Together, these data provide evidence that TAOK2 is a neurodevelopmental disorder risk gene and identify RhoA signaling as a mediator of TAOK2-dependent synaptic development.


Subject(s)
Autism Spectrum Disorder/metabolism , Neurodevelopmental Disorders/metabolism , Protein Serine-Threonine Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Adult , Animals , Anxiety/genetics , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/psychology , Child , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/psychology , Dendrites/metabolism , Dendrites/pathology , Female , Humans , Interpersonal Relations , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Neurodevelopmental Disorders/psychology , Neurogenesis , Phenotype , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Synaptic Transmission , Exome Sequencing
17.
Stem Cell Reports ; 11(5): 1211-1225, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30392976

ABSTRACT

Autism spectrum disorder (ASD) is phenotypically and genetically heterogeneous. We present a CRISPR gene editing strategy to insert a protein tag and premature termination sites creating an induced pluripotent stem cell (iPSC) knockout resource for functional studies of ten ASD-relevant genes (AFF2/FMR2, ANOS1, ASTN2, ATRX, CACNA1C, CHD8, DLGAP2, KCNQ2, SCN2A, TENM1). Neurogenin 2 (NGN2)-directed induction of iPSCs allowed production of excitatory neurons, and mutant proteins were not detectable. RNA sequencing revealed convergence of several neuronal networks. Using both patch-clamp and multi-electrode array approaches, the electrophysiological deficits measured were distinct for different mutations. However, they culminated in a consistent reduction in synaptic activity, including reduced spontaneous excitatory postsynaptic current frequencies in AFF2/FMR2-, ASTN2-, ATRX-, KCNQ2-, and SCN2A-null neurons. Despite ASD susceptibility genes belonging to different gene ontologies, isogenic stem cell resources can reveal common functional phenotypes, such as reduced functional connectivity.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/physiopathology , Gene Editing , Genetic Predisposition to Disease , Neurons/metabolism , Neurons/pathology , Cell Line , Electrodes , Gene Knockout Techniques , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Mutagenesis, Insertional/genetics , Phenotype
18.
Am J Hum Genet ; 102(2): 278-295, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395074

ABSTRACT

Copy-number variations (CNVs) are strong risk factors for neurodevelopmental and psychiatric disorders. The 15q13.3 microdeletion syndrome region contains up to ten genes and is associated with numerous conditions, including autism spectrum disorder (ASD), epilepsy, schizophrenia, and intellectual disability; however, the mechanisms underlying the pathogenesis of 15q13.3 microdeletion syndrome remain unknown. We combined whole-genome sequencing, human brain gene expression (proteome and transcriptome), and a mouse model with a syntenic heterozygous deletion (Df(h15q13)/+ mice) and determined that the microdeletion results in abnormal development of cortical dendritic spines and dendrite outgrowth. Analysis of large-scale genomic, transcriptomic, and proteomic data identified OTUD7A as a critical gene for brain function. OTUD7A was found to localize to dendritic and spine compartments in cortical neurons, and its reduced levels in Df(h15q13)/+ cortical neurons contributed to the dendritic spine and dendrite outgrowth deficits. Our results reveal OTUD7A as a major regulatory gene for 15q13.3 microdeletion syndrome phenotypes that contribute to the disease mechanism through abnormal cortical neuron morphological development.


Subject(s)
Chromosome Disorders/enzymology , Chromosome Disorders/genetics , Deubiquitinating Enzymes/physiology , Endopeptidases/genetics , Intellectual Disability/enzymology , Intellectual Disability/genetics , Neurodevelopmental Disorders/enzymology , Neurodevelopmental Disorders/genetics , Seizures/enzymology , Seizures/genetics , Animals , Autism Spectrum Disorder/genetics , Chromosome Deletion , Chromosomes, Human, Pair 15/enzymology , Chromosomes, Human, Pair 15/genetics , Dendritic Spines/metabolism , Deubiquitinating Enzymes/genetics , Endopeptidases/metabolism , Female , Gene Deletion , Genetic Association Studies , Humans , Male , Mice , Phenotype , Prosencephalon/pathology
19.
J Neurodev Disord ; 8: 45, 2016.
Article in English | MEDLINE | ID: mdl-27980692

ABSTRACT

BACKGROUND: Genetic factors play a major role in the risk for neurodevelopmental disorders such as autism spectrum disorders (ASDs) and intellectual disability (ID). The underlying genetic factors have become better understood in recent years due to advancements in next generation sequencing. These studies have uncovered a vast number of genes that are impacted by different types of mutations (e.g., de novo, missense, truncation, copy number variations). ABSTRACT: Given the large volume of genetic data, analyzing each gene on its own is not a feasible approach and will take years to complete, let alone attempt to use the information to develop novel therapeutics. To make sense of independent genomic data, one approach is to determine whether multiple risk genes function in common signaling pathways that identify signaling "hubs" where risk genes converge. This approach has led to multiple pathways being implicated, such as synaptic signaling, chromatin remodeling, alternative splicing, and protein translation, among many others. In this review, we analyze recent and historical evidence indicating that multiple risk genes, including genes denoted as high-confidence and likely causal, are part of the Wingless (Wnt signaling) pathway. In the brain, Wnt signaling is an evolutionarily conserved pathway that plays an instrumental role in developing neural circuits and adult brain function. CONCLUSIONS: We will also review evidence that pharmacological therapies and genetic mouse models further identify abnormal Wnt signaling, particularly at the synapse, as being disrupted in ASDs and contributing to disease pathology.

20.
Neural Plast ; 2016: 7694385, 2016.
Article in English | MEDLINE | ID: mdl-27847649

ABSTRACT

Cortical inhibitory neurons play crucial roles in regulating excitatory synaptic networks and cognitive function and aberrant development of these cells have been linked to neurodevelopmental disorders. The secreted neurotrophic factor Neuregulin-1 (NRG1) and its receptor ErbB4 are established regulators of inhibitory neuron connectivity, but the developmental signalling mechanisms regulating this process remain poorly understood. Here, we provide evidence that NRG1-ErbB4 signalling functions through the multifunctional scaffold protein, Disrupted in Schizophrenia 1 (DISC1), to regulate the development of cortical inhibitory interneuron dendrite and synaptic growth. We found that NRG1 increases inhibitory neuron dendrite complexity and glutamatergic synapse formation onto inhibitory neurons and that this effect is blocked by expression of a dominant negative DISC1 mutant, or DISC1 knockdown. We also discovered that NRG1 treatment increases DISC1 expression and its localization to glutamatergic synapses being made onto cortical inhibitory neurons. Mechanistically, we determined that DISC1 binds ErbB4 within cortical inhibitory neurons. Collectively, these data suggest that a NRG1-ErbB4-DISC1 signalling pathway regulates the development of cortical inhibitory neuron dendrite and synaptic growth. Given that NRG1, ErbB4, and DISC1 are schizophrenia-linked genes, these findings shed light on how independent risk factors may signal in a common developmental pathway that contributes to neural connectivity defects and disease pathogenesis.


Subject(s)
Cerebral Cortex/physiology , Dendrites/physiology , Nerve Tissue Proteins/biosynthesis , Neuregulin-1/pharmacology , Neurons/physiology , Synapses/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Dendrites/drug effects , HEK293 Cells , Humans , Mice , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Synapses/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...