Parallel in vivo analysis of large-effect autism genes implicates cortical neurogenesis and estrogen in risk and resilience

[1]  H. Willsey,et al.  Xenopus leads the way: Frogs as a pioneering model to understand the human brain , 2020, Genesis.

[2]  Richard A Marini,et al.  Large-scale targeted sequencing identifies risk genes for neurodevelopmental disorders , 2020, Nature Communications.

[3]  M. State,et al.  Neurodevelopmental disorder risk gene DYRK1A is required for ciliogenesis and brain size in Xenopus embryos. , 2020, Development.

[4]  K. Anderson,et al.  Centrosome anchoring regulates progenitor properties and cortical formation , 2020, Nature.

[5]  Matthew W. Mosconi,et al.  Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism , 2019, Cell.

[6]  Laura Pérez-Cano,et al.  Inherited and De Novo Genetic Risk for Autism Impacts Shared Networks , 2019, Cell.

[7]  A. Wills,et al.  Advancing genetic and genomic technologies deepen the pool for discovery in Xenopus tropicalis , 2019, Developmental dynamics : an official publication of the American Association of Anatomists.

[8]  M. Marra,et al.  A distinct neurodevelopmental syndrome with intellectual disability, autism spectrum disorder, characteristic facies, and macrocephaly is caused by defects in CHD8 , 2019, Journal of Human Genetics.

[9]  J. Rosenfeld,et al.  DYRK1A-related intellectual disability: a syndrome associated with congenital anomalies of the kidney and urinary tract , 2019, Genetics in Medicine.

[10]  N. Šestan,et al.  Lost in Translation: Traversing the Complex Path from Genomics to Therapeutics in Autism Spectrum Disorder , 2018, Neuron.

[11]  R. Heald,et al.  Katanin-like protein Katnal2 is required for ciliogenesis and brain development in Xenopus embryos. , 2018, Developmental biology.

[12]  R. Harland,et al.  3D-printable tools for developmental biology: Improving embryo injection and screening techniques through 3D-printing technology , 2018, bioRxiv.

[13]  Nevan J. Krogan,et al.  The Psychiatric Cell Map Initiative: A Convergent Systems Biological Approach to Illuminating Key Molecular Pathways in Neuropsychiatric Disorders , 2018, Cell.

[14]  L. A. Lowery,et al.  The Role of the Microtubule Cytoskeleton in Neurodevelopmental Disorders , 2018, Front. Cell. Neurosci..

[15]  Wei Zhang,et al.  Systematic Evaluation of Molecular Networks for Discovery of Disease Genes. , 2018, Cell systems.

[16]  Joseph G Ibrahim,et al.  Heavy-tailed prior distributions for sequence count data: removing the noise and preserving large differences , 2018, bioRxiv.

[17]  N. Zečević,et al.  The Subventricular Zone: A Key Player in Human Neocortical Development , 2018, The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry.

[18]  Emily K. Mis,et al.  CRISPR/Cas9 F0 Screening of Congenital Heart Disease Genes in Xenopus tropicalis. , 2018, Methods in molecular biology.

[19]  R. Mikut,et al.  Tracking of Indels by DEcomposition is a Simple and Effective Method to Assess Efficiency of Guide RNAs in Zebrafish. , 2017, Zebrafish.

[20]  M. Horb,et al.  Tissue-Specific Gene Inactivation in Xenopus laevis: Knockout of lhx1 in the Kidney with CRISPR/Cas9 , 2017, Genetics.

[21]  Yufeng Shen,et al.  Contribution of rare inherited and de novo variants in 2,871 congenital heart disease probands , 2017, Nature Genetics.

[22]  Yong-hui Jiang,et al.  Altered neurogenesis and disrupted expression of synaptic proteins in prefrontal cortex of SHANK3-deficient non-human primate , 2017, Cell Research.

[23]  M. Khokha,et al.  An Interspecies Heart-to-Heart: Using Xenopus to Uncover the Genetic Basis of Congenital Heart Disease , 2017, Current Pathobiology Reports.

[24]  A. McMahon,et al.  Hedgehog Signaling: From Basic Biology to Cancer Therapy. , 2017, Cell chemical biology.

[25]  A. Brivanlou,et al.  Combined small-molecule inhibition accelerates the derivation of functional, early-born, cortical neurons from human pluripotent stem cells , 2017, Nature Biotechnology.

[26]  J. Thornton,et al.  Structural analysis of pathogenic mutations in the DYRK1A gene in patients with developmental disorders , 2017, Human molecular genetics.

[27]  B. DeLay,et al.  Xenopus: leaping forward in kidney organogenesis , 2017, Pediatric Nephrology.

[28]  M. Suyama,et al.  CHD8 haploinsufficiency results in autistic-like phenotypes in mice , 2016, Nature.

[29]  Max A. Horlbeck,et al.  Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation , 2016, eLife.

[30]  Carol Y. B. Liu,et al.  Chd8 mediates cortical neurogenesis via transcriptional regulation of cell cycle and Wnt signaling , 2016, Nature Neuroscience.

[31]  A. Packer Neocortical neurogenesis and the etiology of autism spectrum disorder , 2016, Neuroscience & Biobehavioral Reviews.

[32]  Nevan J Krogan,et al.  CRISPR Interference Efficiently Induces Specific and Reversible Gene Silencing in Human iPSCs. , 2016, Cell stem cell.

[33]  M. Khokha,et al.  Xenopus as a model organism for birth defects-Congenital heart disease and heterotaxy. , 2016, Seminars in cell & developmental biology.

[34]  Joseph M. Fernandez,et al.  Estrogens Suppress a Behavioral Phenotype in Zebrafish Mutants of the Autism Risk Gene, CNTNAP2 , 2016, Neuron.

[35]  Kali T. Witherspoon,et al.  Disruptive de novo mutations of DYRK1A lead to a syndromic form of autism and ID , 2016, Molecular Psychiatry.

[36]  Maura Lane,et al.  CRISPR/Cas9: An inexpensive, efficient loss of function tool to screen human disease genes in Xenopus. , 2015, Developmental biology.

[37]  Zhihai Ma,et al.  Identification of Human Neuronal Protein Complexes Reveals Biochemical Activities and Convergent Mechanisms of Action in Autism Spectrum Disorders. , 2015, Cell systems.

[38]  Joshua L. Deignan,et al.  DYRK1A haploinsufficiency causes a new recognizable syndrome with microcephaly, intellectual disability, speech impairment, and distinct facies , 2015, European Journal of Human Genetics.

[39]  W. Chung,et al.  De novo POGZ mutations are associated with neurodevelopmental disorders and microcephaly , 2015, Cold Spring Harbor molecular case studies.

[40]  Christopher S. Poultney,et al.  Insights into Autism Spectrum Disorder Genomic Architecture and Biology from 71 Risk Loci , 2015, Neuron.

[41]  Charles E. Vejnar,et al.  CRISPRscan: designing highly efficient sgRNAs for CRISPR/Cas9 targeting in vivo , 2015, Nature Methods.

[42]  D. Geschwind,et al.  Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture , 2015, Nature Methods.

[43]  Zengqiang Yuan,et al.  CHD2 is Required for Embryonic Neurogenesis in the Developing Cerebral Cortex , 2015, Stem cells.

[44]  Matthew W State,et al.  Autism spectrum disorders: from genes to neurobiology , 2015, Current Opinion in Neurobiology.

[45]  Mahmood Rasool,et al.  Molecular genetics of human primary microcephaly: an overview , 2015, BMC Medical Genomics.

[46]  Stephan J. Sanders,et al.  Genotype to phenotype relationships in autism spectrum disorders , 2014, Nature Neuroscience.

[47]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[48]  Boris Yamrom,et al.  The contribution of de novo coding mutations to autism spectrum disorder , 2014, Nature.

[49]  Christopher S. Poultney,et al.  Synaptic, transcriptional, and chromatin genes disrupted in autism , 2014, Nature.

[50]  B. van Steensel,et al.  Easy quantitative assessment of genome editing by sequence trace decomposition , 2014, Nucleic acids research.

[51]  Yoav Gilad,et al.  A panel of induced pluripotent stem cells from chimpanzees: a resource for comparative functional genomics , 2014, bioRxiv.

[52]  Jay Shendure,et al.  Disruptive CHD8 Mutations Define a Subtype of Autism Early in Development , 2014, Cell.

[53]  Allan R. Jones,et al.  Transcriptional Landscape of the Prenatal Human Brain , 2014, Nature.

[54]  P. Krieg,et al.  Reagents for developmental regulation of Hedgehog signaling. , 2014, Methods.

[55]  N. Šestan,et al.  The developmental transcriptome of the human brain: implications for neurodevelopmental disorders. , 2014, Current opinion in neurology.

[56]  David R. O'Brien,et al.  Cell Type-Specific Expression Analysis to Identify Putative Cellular Mechanisms for Neurogenetic Disorders , 2014, The Journal of Neuroscience.

[57]  Luke A. Gilbert,et al.  Dynamic Imaging of Genomic Loci in Living Human Cells by an Optimized CRISPR/Cas System , 2013, Cell.

[58]  B. Conklin,et al.  Induced pluripotent stem cells from patients with human fibrodysplasia ossificans progressiva show increased mineralization and cartilage formation , 2013, Orphanet Journal of Rare Diseases.

[59]  Wei Niu,et al.  Coexpression Networks Implicate Human Midfetal Deep Cortical Projection Neurons in the Pathogenesis of Autism , 2013, Cell.

[60]  S. Horvath,et al.  Integrative Functional Genomic Analyses Implicate Specific Molecular Pathways and Circuits in Autism , 2013, Cell.

[61]  E. Ben-David,et al.  Combined analysis of exome sequencing points toward a major role for transcription regulation during brain development in autism , 2013, Molecular Psychiatry.

[62]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[63]  Bradley P. Coe,et al.  Multiplex Targeted Sequencing Identifies Recurrently Mutated Genes in Autism Spectrum Disorders , 2012, Science.

[64]  Matthew W. State,et al.  The Emerging Biology of Autism Spectrum Disorders , 2012, Science.

[65]  Johannes E. Schindelin,et al.  Fiji: an open-source platform for biological-image analysis , 2012, Nature Methods.

[66]  Kenny Q. Ye,et al.  De Novo Gene Disruptions in Children on the Autistic Spectrum , 2012, Neuron.

[67]  Michael F. Walker,et al.  De novo mutations revealed by whole-exome sequencing are strongly associated with autism , 2012, Nature.

[68]  Evan T. Geller,et al.  Patterns and rates of exonic de novo mutations in autism spectrum disorders , 2012, Nature.

[69]  M. Horb,et al.  Development of xenopus resource centers: The national xenopus resource and the european xenopus resource center , 2012, Genesis.

[70]  Bradley P. Coe,et al.  Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations , 2012, Nature.

[71]  Pat Levitt,et al.  The conundrums of understanding genetic risks for autism spectrum disorders , 2011, Nature Neuroscience.

[72]  J. Kleinman,et al.  Spatiotemporal transcriptome of the human brain , 2011, Nature.

[73]  Diana V. Dugas,et al.  Protein Interactome Reveals Converging Molecular Pathways Among Autism Disorders , 2011, Science Translational Medicine.

[74]  Nobutoshi Ito,et al.  Development of a novel selective inhibitor of the Down syndrome-related kinase Dyrk1A. , 2010, Nature communications.

[75]  Anushya Muruganujan,et al.  PANTHER version 7: improved phylogenetic trees, orthologs and collaboration with the Gene Ontology Consortium , 2009, Nucleic Acids Res..

[76]  W. Becker,et al.  Harmine specifically inhibits protein kinase DYRK1A and interferes with neurite formation , 2009, The FEBS journal.

[77]  T. Pieler,et al.  Comparative expression analysis of the neurogenins in Xenopus tropicalis and Xenopus laevis , 2009, Developmental dynamics : an official publication of the American Association of Anatomists.

[78]  G. Fried,et al.  Transcriptional Analysis of Estrogen Effects in Human Embryonic Neurons and Glial Cells , 2008, Neuroendocrinology.

[79]  K. Shiota,et al.  Hedgehog signaling is involved in development of the neocortex , 2008, Development.

[80]  R. Harland,et al.  Baskets for in situ hybridization and immunohistochemistry. , 2007, CSH protocols.

[81]  T. Iguchi,et al.  Molecular cloning of two isoforms of Xenopus (Silurana) tropicalis estrogen receptor mRNA and their expression during development. , 2007, Biochimica et biophysica acta.

[82]  A. Kriegstein,et al.  Patterns of neural stem and progenitor cell division may underlie evolutionary cortical expansion , 2006, Nature Reviews Neuroscience.

[83]  A. Kriegstein,et al.  The role of intermediate progenitor cells in the evolutionary expansion of the cerebral cortex. , 2006, Cerebral cortex.

[84]  Sarah Barber,et al.  Sequencing and analysis of 10,967 full-length cDNA clones from Xenopus laevis and Xenopus tropicalis reveals post-tetraploidization transcriptome remodeling. , 2006, Genome research.

[85]  N. Zečević,et al.  Contributions of cortical subventricular zone to the development of the human cerebral cortex , 2005, The Journal of comparative neurology.

[86]  G. Fishell,et al.  Temporal requirement for hedgehog signaling in ventral telencephalic patterning , 2004, Development.

[87]  D. Kelley,et al.  Estrogen receptors in Xenopus: duplicate genes, splice variants, and tissue-specific expression. , 2003, General and comparative endocrinology.

[88]  Jussi Taipale,et al.  Inhibition of Hedgehog signaling by direct binding of cyclopamine to Smoothened. , 2002, Genes & development.

[89]  Y. Ohkubo,et al.  Coordinate regulation and synergistic actions of BMP4, SHH and FGF8 in the rostral prosencephalon regulate morphogenesis of the telencephalic and optic vesicles , 2002, Neuroscience.

[90]  R. Keynes,et al.  A critical role for sonic hedgehog signaling in the early expansion of the developing brain , 2002, Nature Neuroscience.

[91]  Alex E. Lash,et al.  Gene Expression Omnibus: NCBI gene expression and hybridization array data repository , 2002, Nucleic Acids Res..

[92]  T. Pieler,et al.  Molecular cloning and expression analysis of the Hedgehog receptors XPtc1 and XSmo in Xenopus laevis , 2001, Mechanisms of Development.

[93]  J. Rubenstein,et al.  Inductive interactions direct early regionalization of the mouse forebrain. , 1997, Development.

[94]  S. Anderson,et al.  Mutations of the Homeobox Genes Dlx-1 and Dlx-2 Disrupt the Striatal Subventricular Zone and Differentiation of Late Born Striatal Neurons , 1997, Neuron.

[95]  Andrew P. McMahon,et al.  The world according to bedgebog , 1997 .

[96]  T. Jessell,et al.  Graded sonic hedgehog signaling and the specification of cell fate in the ventral neural tube. , 1997, Cold Spring Harbor symposia on quantitative biology.

[97]  T. Jessell,et al.  Diversity and Pattern in the Developing Spinal Cord , 1996, Science.

[98]  P. Beachy,et al.  Cyclopia and defective axial patterning in mice lacking Sonic hedgehog gene function , 1996, Nature.

[99]  T. Lints,et al.  Sonic hedgehog induces the differentiation of ventral forebrain neurons: A common signal for ventral patterning within the neural tube , 1995, Cell.

[100]  Andrew P. McMahon,et al.  Sonic hedgehog, a member of a family of putative signaling molecules, is implicated in the regulation of CNS polarity , 1993, Cell.

[101]  R. Harland,et al.  In situ hybridization: an improved whole-mount method for Xenopus embryos. , 1991, Methods in cell biology.