51
|
Lee Y, Han PL. Early-Life Stress in D2 Heterozygous Mice Promotes Autistic-like Behaviors through the Downregulation of the BDNF-TrkB Pathway in the Dorsal Striatum. Exp Neurobiol 2019; 28:337-351. [PMID: 31308794 PMCID: PMC6614072 DOI: 10.5607/en.2019.28.3.337] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/30/2019] [Accepted: 06/05/2019] [Indexed: 12/12/2022] Open
Abstract
A number of specific genetic variants including gene mutations and single nucleotide variations have been identified in genomewide association studies of autism spectrum disorder (ASD). ASD phenotypes in individuals carrying specific genetic variations are manifest mostly in a heterozygous state. Furthermore, individuals with most genetic variants show incomplete penetrance and phenotypic variability, suggesting that non-genetic factors are also involved in developing ASD. However, the mechanisms of how genetic and environmental factors interactively promote ASD are not clearly understood. In the present study, we investigated whether early-life stress (ELS) in D2 dopamine receptor heterozygous knockout (D2+/−) mice induces ASD-like symptoms. To address that, we exposed D2 heterozygous pups to maternal separation stress for 3 h daily for 13 days beginning on postnatal day 2. D2+/− adult mice that had experienced ELS exhibited impaired sociability in the three-chamber test and home-cage social interaction test and increased grooming behavior, whereas wildtype littermates exposed to ELS did not show those phenotypes. ELS-exposed D2+/− mice had decreased levels of BDNF, TrkB, phospho-ERK1/2 and phospho-CREB in the dorsal striatum. Administration of the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) to ELS-exposed D2+/− mice rescued the sociability deficits and repetitive behavior. In contrast, behavioral rescue by 7,8-DHF in ELS-exposed D2+/− mice was blocked when TrkB expression in the dorsal striatum was locally inhibited by the injection of TrkB-siRNA. Together, our results suggest that the interaction between ELS and defective D2 gene function promotes autistic-like behaviors by downregulating the BDNF-TrkB pathway in the dorsal striatum.
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Pyung-Lim Han
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea.,Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
52
|
Sullivan JM, De Rubeis S, Schaefer A. Convergence of spectrums: neuronal gene network states in autism spectrum disorder. Curr Opin Neurobiol 2019; 59:102-111. [PMID: 31220745 DOI: 10.1016/j.conb.2019.04.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/20/2019] [Accepted: 04/24/2019] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder characterized by social deficits and restrictive and/or repetitive behaviors. The breadth of ASD symptoms is paralleled by the multiplicity of genes that have been implicated in its etiology. Initial findings revealed numerous ASD risk genes that contribute to synaptic function. More recently, genomic and gene expression studies point to altered chromatin function and impaired transcriptional control as additional risk factors for ASD. The consequences of impaired transcriptional alterations in ASD involve consistent changes in synaptic gene expression and cortical neuron specification during brain development. The multiplicity of genetic and environmental factors associated with ASD risk and their convergence onto common molecular pathways in neurons point to ASD as a disorder of gene regulatory networks.
Collapse
Affiliation(s)
- Josefa M Sullivan
- Nash Family Department of Neuroscience, New York, NY, USA; Department of Psychiatry, New York, NY, USA; Friedman Brain Institute, New York, NY, USA; Seaver Autism Center for Research and Treatment, New York, NY, USA
| | - Silvia De Rubeis
- Department of Psychiatry, New York, NY, USA; Seaver Autism Center for Research and Treatment, New York, NY, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anne Schaefer
- Nash Family Department of Neuroscience, New York, NY, USA; Department of Psychiatry, New York, NY, USA; Friedman Brain Institute, New York, NY, USA; Seaver Autism Center for Research and Treatment, New York, NY, USA.
| |
Collapse
|
53
|
Foxp1 is critical for the maintenance of regulatory T-cell homeostasis and suppressive function. PLoS Biol 2019; 17:e3000270. [PMID: 31125332 PMCID: PMC6534289 DOI: 10.1371/journal.pbio.3000270] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 04/30/2019] [Indexed: 12/13/2022] Open
Abstract
Regulatory T (Treg) cells play central roles in maintaining immune homeostasis and self-tolerance. However, the molecular mechanisms underlying Treg cell homeostasis and suppressive function are still not fully understood. Here, we report that the deletion of another P subfamily members of the forkhead box (Foxp) subfamily member Foxp1 in Treg cells led to increased numbers of activated Treg (aTreg) cells at the expense of quiescent Treg cells, and also resulted in impaired Treg suppressive function. Mice with Foxp1-deficient Treg cells developed spontaneous inflammatory disease with age; they also had more severe inflammatory disease in colitis and experimental autoimmune encephalomyelitis (EAE) models. Mechanistically, we found that Foxp1 bound to the conserved noncoding sequence 2 (CNS2) element of the Foxp3 locus and helped maintain Treg suppressive function by stabilizing the Foxp3 expression. Furthermore, we found that Foxp1 and Foxp3 coordinated the regulation of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) expression levels. Taken together, our study demonstrates that Foxp1 plays critical roles in both maintaining Treg cell quiescence during homeostasis and regulating Treg suppressive function. The transcription factor Foxp3 has been considered as the master regulator of regulatory T cells (Tregs), but this study shows that another Foxp subfamily member, Foxp1, plays important roles in the homeostasis, stability, and suppressor function of Tregs.
Collapse
|
54
|
Jay K, Mitra A, Harding T, Matthes D, Van Ness B. Identification of a de novo FOXP1 mutation and incidental discovery of inherited genetic variants contributing to a case of autism spectrum disorder and epilepsy. Mol Genet Genomic Med 2019; 7:e00751. [PMID: 31111659 PMCID: PMC6625142 DOI: 10.1002/mgg3.751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/08/2019] [Accepted: 04/22/2019] [Indexed: 12/15/2022] Open
Abstract
Background Autism spectrum disorder is commonly co‐diagnosed intellectual disability, language disorder, anxiety, and epilepsy, however, symptom management is difficult due to the complex genetic nature of ASD. Methods We present a next‐generation sequencing‐based case study with both de novo and inherited genetic variants and highlight the impact of structural variants on post‐translational regulation of protein expression. Since management of symptoms has classically been through pharmaceutical therapies, a pharmacogenomics screen was also utilized to determine possible drug/gene interactions. Results A de novo variant was identified within the FOXP1 3′ untranslated regulatory region using exome sequencing. Additionally, inherited variants that likely contribute to the current and potential future traits were identified within the COMT, SLC6A4, CYP2C19, and CYP2D6 genes. Conclusion This study aims to elucidate how a collection of variant genotypes could potentially impact neural development resulting in a unique phenotype including ASD and epilepsy. Each gene's contribution to neural development is assessed, and the interplay of these genotypes is discussed. The results highlight the utility of exome sequencing in conjunction with pharmacogenomics screening when evaluating possible causes of and therapeutic treatments for ASD‐related symptoms.
Collapse
Affiliation(s)
- Kristy Jay
- College of Biological Sciences, Department of Genetics, Cell Biology, and Development, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Amit Mitra
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama
| | - Taylor Harding
- College of Biological Sciences, Department of Genetics, Cell Biology, and Development, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - David Matthes
- College of Biological Sciences, Department of Biology, Teaching, and Learning, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Brian Van Ness
- College of Biological Sciences, Department of Genetics, Cell Biology, and Development, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| |
Collapse
|
55
|
Ayhan F, Konopka G. Regulatory genes and pathways disrupted in autism spectrum disorders. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:57-64. [PMID: 30165121 PMCID: PMC6249101 DOI: 10.1016/j.pnpbp.2018.08.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/01/2018] [Accepted: 08/21/2018] [Indexed: 02/07/2023]
Abstract
Autism spectrum disorder (ASD) is a highly prevalent and complex genetic disorder. The complex genetic make-up of ASD has been extensively studied and both common and rare genetic variants in up to 1000 genes have been linked to increased ASD risk. While these studies highlight the genetic complexity and begin to provide a window for delineating pathways at risk in ASD, the pathogenicity and specific contribution of many mutations to the disorder are poorly understood. Defining the convergent pathways disrupted by this large number of ASD-associated genetic variants will help to understand disease pathogenesis and direct future therapeutic efforts for the groups of patients with distinct etiologies. Here, we review some of the common regulatory pathways including chromatin remodeling, transcription, and alternative splicing that have emerged as common features from genetic and transcriptomic profiling of ASD. For each category, we focus on one gene (CHD8, FOXP1, and RBFOX1) that is significantly linked to ASD and functionally characterized in recent years. Finally, we discuss genetic and transcriptomic overlap between ASD and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Fatma Ayhan
- Department of Neuroscience, UT Southwestern Medical Center, Dallas 75390-9111, USA
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas 75390-9111, USA.
| |
Collapse
|
56
|
Castells-Nobau A, Eidhof I, Fenckova M, Brenman-Suttner DB, Scheffer-de Gooyert JM, Christine S, Schellevis RL, van der Laan K, Quentin C, van Ninhuijs L, Hofmann F, Ejsmont R, Fisher SE, Kramer JM, Sigrist SJ, Simon AF, Schenck A. Conserved regulation of neurodevelopmental processes and behavior by FoxP in Drosophila. PLoS One 2019; 14:e0211652. [PMID: 30753188 PMCID: PMC6372147 DOI: 10.1371/journal.pone.0211652] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/30/2022] Open
Abstract
FOXP proteins form a subfamily of evolutionarily conserved transcription factors involved in the development and functioning of several tissues, including the central nervous system. In humans, mutations in FOXP1 and FOXP2 have been implicated in cognitive deficits including intellectual disability and speech disorders. Drosophila exhibits a single ortholog, called FoxP, but due to a lack of characterized mutants, our understanding of the gene remains poor. Here we show that the dimerization property required for mammalian FOXP function is conserved in Drosophila. In flies, FoxP is enriched in the adult brain, showing strong expression in ~1000 neurons of cholinergic, glutamatergic and GABAergic nature. We generate Drosophila loss-of-function mutants and UAS-FoxP transgenic lines for ectopic expression, and use them to characterize FoxP function in the nervous system. At the cellular level, we demonstrate that Drosophila FoxP is required in larvae for synaptic morphogenesis at axonal terminals of the neuromuscular junction and for dendrite development of dorsal multidendritic sensory neurons. In the developing brain, we find that FoxP plays important roles in α-lobe mushroom body formation. Finally, at a behavioral level, we show that Drosophila FoxP is important for locomotion, habituation learning and social space behavior of adult flies. Our work shows that Drosophila FoxP is important for regulating several neurodevelopmental processes and behaviors that are related to human disease or vertebrate disease model phenotypes. This suggests a high degree of functional conservation with vertebrate FOXP orthologues and established flies as a model system for understanding FOXP related pathologies.
Collapse
Affiliation(s)
- Anna Castells-Nobau
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ilse Eidhof
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Michaela Fenckova
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Jolanda M. Scheffer-de Gooyert
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sheren Christine
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rosa L. Schellevis
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kiran van der Laan
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Christine Quentin
- Genetics, Institute of Biology, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Lisa van Ninhuijs
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Falko Hofmann
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Radoslaw Ejsmont
- Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Dresden, Germany
| | - Simon E. Fisher
- Language and Genetics Department, Max Planck Institute of Psycholinguistics, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Jamie M. Kramer
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Stephan J. Sigrist
- Genetics, Institute of Biology, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Anne F. Simon
- Department of Biology, Faculty of Science, Western University, London, Ontario, Canada
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
57
|
Xu H, Liu F. Downregulation of FOXP1 correlates with tendon stem/progenitor cells aging. Biochem Biophys Res Commun 2018; 504:96-102. [PMID: 30170733 DOI: 10.1016/j.bbrc.2018.08.136] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/22/2018] [Indexed: 12/18/2022]
Abstract
Aging is known as a major risk factor for tendon disorders whereas the molecular mechanisms of age-related tendon disorders still remains unclear. Since tendon-derived stem/progenitor cells (TSPCs) play a vital role in tendon maintenance and healing, in this study we aimed to investigate the role of Forkhead box P1 (FOXP1) in aged TSPCs, we found that FOXP1 mRNA and protein levels were markedly decreased in the aged TSPCs. Moreover, overexpression of FOXP1 attenuates TSPCs aging, as confirmed by decreased of senescence-associated β-gal staining, as well as the senescence marker, p16INK4A. Conversely, FOXP1 depletion by siRNA promoted senescence in young TSPCs. Meanwhile, FOXP1 overexpression also restores the age-associated reduction of self-renewal, migration and differentiation of TSPCs. In addition, FOXP1 overexpression rescued decreased levels of E2F1, pRb and cyclin D1 in aged TSPCs, which suggested that FOXP1 regulates TSPCs aging through cellular senescence. These results indicate that FOXP1 plays a crucial role in TSPCs aging.
Collapse
Affiliation(s)
- Hua Xu
- First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China; Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Fan Liu
- First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China; Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China.
| |
Collapse
|
58
|
Braccioli L, Vervoort SJ, Adolfs Y, Heijnen CJ, Basak O, Pasterkamp RJ, Nijboer CH, Coffer PJ. FOXP1 Promotes Embryonic Neural Stem Cell Differentiation by Repressing Jagged1 Expression. Stem Cell Reports 2018; 9:1530-1545. [PMID: 29141232 PMCID: PMC5688236 DOI: 10.1016/j.stemcr.2017.10.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 01/11/2023] Open
Abstract
Mutations in FOXP1 have been linked to neurodevelopmental disorders including intellectual disability and autism; however, the underlying molecular mechanisms remain ill-defined. Here, we demonstrate with RNA and chromatin immunoprecipitation sequencing that FOXP1 directly regulates genes controlling neurogenesis. We show that FOXP1 is expressed in embryonic neural stem cells (NSCs), and modulation of FOXP1 expression affects both neuron and astrocyte differentiation. Using a murine model of cortical development, FOXP1-knockdown in utero was found to reduce NSC differentiation and migration during corticogenesis. Furthermore, transplantation of FOXP1-knockdown NSCs in neonatal mice after hypoxia-ischemia challenge demonstrated that FOXP1 is also required for neuronal differentiation and functionality in vivo. FOXP1 was found to repress the expression of Notch pathway genes including the Notch-ligand Jagged1, resulting in inhibition of Notch signaling. Finally, blockade of Jagged1 in FOXP1-knockdown NSCs rescued neuronal differentiation in vitro. Together, these data support a role for FOXP1 in regulating embryonic NSC differentiation by modulating Notch signaling. FOXP1 promotes astrocyte and neuronal differentiation of NSCs in vitro FOXP1 promotes neuronal differentiation of NSCs in vivo FOXP1 transcriptionally regulates pro-neural genes and represses Notch pathway genes FOXP1 promotes neuronal differentiation by limiting Jagged1 expression
Collapse
Affiliation(s)
- Luca Braccioli
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands; Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands
| | - Stephin J Vervoort
- Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, the Netherlands
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Onur Basak
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, the Netherlands
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands.
| | - Paul J Coffer
- Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands.
| |
Collapse
|
59
|
A genome scan for quantitative trait loci affecting average daily gain and Kleiber ratio in Baluchi Sheep. J Genet 2018. [DOI: 10.1007/s12041-018-0941-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
60
|
Pasandideh M, Rahimi-Mianji G, Gholizadeh M. A genome scan for quantitative trait loci affecting average daily gain and Kleiber ratio in Baluchi Sheep. J Genet 2018; 97:493-503. [PMID: 29932070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Genomewide association study (GWAS) is an efficient tool for the detection of SNPs and candidate genes in quantitative traits. Growth rate is an important trait for increasing the meat production in sheep. A total of 96 Baluchi sheep were genotyped using Illumina Ovine SNP50 BeadChip to run a GWAS for an average daily gain (ADG) and Kleiber ratio (KR) traits in different periods of age in sheep. Traits included were average daily gain from birth to three months (ADG0-3), from three months to six months (ADG3-6), from six months to nine months (ADG6-9), from nine months to yearling (ADG9-12), from birth to six months (ADG0-6), from three months to nine months (ADG3-9), from three months to yearling (ADG3-12) and corresponding Kleiber ratios (KR0-3, KR3-6, KR6-9, KR9-12, KR0-6, KR3-9 and KR3-12, respectively). A total of 42,243 SNPs passed the quality-control filters and were analysed by PLINK software in a linear mixed model. Two SNPs were identified on two chromosomes at the 5% genomewide significance level for KR(3-9) and KR(6-9). Two candidate genes, namely MAGI1 and ZNF770, were identified correspondingly harbouring and close to these QTL. Also, a total of 21 SNPs were found on chromosomes 2, 3, 5, 6, 7, 10, 17, 19, 20 and 25 at the 5% chromosomewide significance level for ADG and KR traits. Thus, we suggest more studies to discover the causative variants for growth traits in sheep.
Collapse
Affiliation(s)
- Majid Pasandideh
- Laboratory for Molecular Genetics and Animal Biotechnology, Faculty of Animal and Aquatic Science, Sari Agricultural Sciences and Natural Resources University, P.O. Box 578, Sari, Iran.
| | | | | |
Collapse
|
61
|
Goin-Kochel RP, Trinh S, Barber S, Bernier R. Gene Disrupting Mutations Associated with Regression in Autism Spectrum Disorder. J Autism Dev Disord 2018; 47:3600-3607. [PMID: 28856484 DOI: 10.1007/s10803-017-3256-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Approximately one-third of children with autism spectrum disorder (ASD) reportedly lose skills within the first 3 years, yet a causal mechanism remains elusive. Considering evidence of strong genetic effects for ASD and findings that distinct phenotypes in ASD associate with specific genetic events, we examined rates of parent-reported regression in the Simons Simplex Collection with likely gene disrupting mutations from five distinct classes: FMRP target genes, genes encoding chromatin modifiers, genes expressed preferentially in embryos, genes encoding postsynaptic density proteins, and essential genes. Children with ASD and mutations in postsynaptic density genes were more likely to experience regression, while a trend suggested that children with ASD and mutations in embryonic genes were less likely to have skill losses.
Collapse
Affiliation(s)
- Robin P Goin-Kochel
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA. .,Autism Center, Texas Children's Hospital, Houston, TX, USA.
| | - Sandy Trinh
- Department of School Psychology, University of Washington, Seattle, Washington, USA
| | - Shelley Barber
- Department of School Psychology, University of Washington, Seattle, Washington, USA
| | - Raphael Bernier
- Department of Psychiatry, University of Washington, Seattle, Washington, USA. .,Center on Human Development and Disability, University of Washington, Seattle, WA, USA.
| |
Collapse
|
62
|
Donizy P, Pagacz K, Marczuk J, Fendler W, Maciejczyk A, Halon A, Matkowski R. Upregulation of FOXP1 is a new independent unfavorable prognosticator and a specific predictor of lymphatic dissemination in cutaneous melanoma patients. Onco Targets Ther 2018; 11:1413-1422. [PMID: 29559799 PMCID: PMC5857151 DOI: 10.2147/ott.s151286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background FOXP1 is a pleiotropic protein that plays important roles in immune responses (B-cell development regulation and differentiation of monocyte), organ development (cardiac valves, lung, and esophagus), and neuronal development. Besides being the primary regulator of normal human tissue development, FOXP1 also plays a role in tumorigenesis. However, the potential value of FOXP1 expression in tumor prognosis remains controversial. FOXP1 expression was assessed in tumor cells (TCs) and stromal cells (SCs) of cutaneous melanomas with the aim of analyzing the associations between FOXP1 expression and clinicopathological characteristics. We believe this article to be the first report analyzing the correlations between FOXP1 expression and clinicopathological, as well as histological, characteristics in melanoma. Materials and methods In total, 96 formalin-fixed, paraffin-embedded primary cutaneous melanoma tissue specimens were subjected to immunohistochemical analysis for FOXP1, and the results were correlated with classical clinicopathological features and patient survival. Results FOXP1 overexpression in TCs was strongly associated with the presence of metastases in sentinel lymph nodes (p=0.0003, OR=11.66) and positive status of regional lymph nodes (p=0.0006, OR=22.15). In 96% (52 of 54) of patients presenting with low FOXP1 expression, no clinical or histopathological features of lymphatic dissemination were observed. However, thinner and nonulcerated tumors were reported to have increased numbers of FOXP1-positive SCs. In addition, a strong association was observed between FOXP1 upregulation in SCs and the absence of regional lymph node metastases. There was a significant correlation between FOXP1 upregulation in TCs and shorter cancer-specific overall survival (log-rank test, p=0.0040) and disease-free survival (log-rank test, p=0.0021). FOXP1 expression was confirmed in multivariate analysis as a factor that significantly unfavorably impacts prognosis in melanoma patients (HR=3.14, p=0.0299, adjusted for age, Breslow thickness, and sex). Conclusion The findings from this study indicate that FOXP1 has a major role in melanoma progression, which makes it a candidate for molecular target-based cancer therapy.
Collapse
Affiliation(s)
- Piotr Donizy
- Department of Pathomorphology and Oncological Cytology, Wroclaw Medical University, Wroclaw, Poland
| | - Konrad Pagacz
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Jakub Marczuk
- Department of Pathomorphology and Oncological Cytology, Wroclaw Medical University, Wroclaw, Poland
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Adam Maciejczyk
- Department of Oncology and Clinic of Radiation Oncology, Wroclaw Medical University, Wroclaw, Poland.,Lower Silesian Oncology Centre, Wroclaw, Poland
| | - Agnieszka Halon
- Department of Pathomorphology and Oncological Cytology, Wroclaw Medical University, Wroclaw, Poland
| | - Rafal Matkowski
- Lower Silesian Oncology Centre, Wroclaw, Poland.,Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
63
|
Rodenas-Cuadrado PM, Mengede J, Baas L, Devanna P, Schmid TA, Yartsev M, Firzlaff U, Vernes SC. Mapping the distribution of language related genes FoxP1, FoxP2, and CntnaP2 in the brains of vocal learning bat species. J Comp Neurol 2018; 526:1235-1266. [PMID: 29297931 PMCID: PMC5900884 DOI: 10.1002/cne.24385] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/07/2017] [Accepted: 11/27/2017] [Indexed: 11/17/2022]
Abstract
Genes including FOXP2, FOXP1, and CNTNAP2, have been implicated in human speech and language phenotypes, pointing to a role in the development of normal language‐related circuitry in the brain. Although speech and language are unique to humans a comparative approach is possible by addressing language‐relevant traits in animal systems. One such trait, vocal learning, represents an essential component of human spoken language, and is shared by cetaceans, pinnipeds, elephants, some birds and bats. Given their vocal learning abilities, gregarious nature, and reliance on vocalizations for social communication and navigation, bats represent an intriguing mammalian system in which to explore language‐relevant genes. We used immunohistochemistry to detail the distribution of FoxP2, FoxP1, and Cntnap2 proteins, accompanied by detailed cytoarchitectural histology in the brains of two vocal learning bat species; Phyllostomus discolor and Rousettus aegyptiacus. We show widespread expression of these genes, similar to what has been previously observed in other species, including humans. A striking difference was observed in the adult P. discolor bat, which showed low levels of FoxP2 expression in the cortex that contrasted with patterns found in rodents and nonhuman primates. We created an online, open‐access database within which all data can be browsed, searched, and high resolution images viewed to single cell resolution. The data presented herein reveal regions of interest in the bat brain and provide new opportunities to address the role of these language‐related genes in complex vocal‐motor and vocal learning behaviors in a mammalian model system.
Collapse
Affiliation(s)
- Pedro M Rodenas-Cuadrado
- Neurogenetics of Vocal Communication Group, Max Planck Institute for Psycholinguistics, Nijmegen, 6500 AH, The Netherlands
| | - Janine Mengede
- Neurogenetics of Vocal Communication Group, Max Planck Institute for Psycholinguistics, Nijmegen, 6500 AH, The Netherlands
| | - Laura Baas
- Neurogenetics of Vocal Communication Group, Max Planck Institute for Psycholinguistics, Nijmegen, 6500 AH, The Netherlands
| | - Paolo Devanna
- Neurogenetics of Vocal Communication Group, Max Planck Institute for Psycholinguistics, Nijmegen, 6500 AH, The Netherlands
| | - Tobias A Schmid
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, California, 94720
| | - Michael Yartsev
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, California, 94720.,Department of Bioengineering, UC Berkeley, 306 University of California, Berkeley, California, 94720
| | - Uwe Firzlaff
- Department Tierwissenschaften, Lehrstuhl für Zoologie, TU München, München, 85354, Germany
| | - Sonja C Vernes
- Neurogenetics of Vocal Communication Group, Max Planck Institute for Psycholinguistics, Nijmegen, 6500 AH, The Netherlands.,Donders Centre for Cognitive Neuroimaging, Nijmegen, 6525 EN, The Netherlands
| |
Collapse
|
64
|
Urreizti R, Damanti S, Esteve C, Franco-Valls H, Castilla-Vallmanya L, Tonda R, Cormand B, Vilageliu L, Opitz JM, Neri G, Grinberg D, Balcells S. A De Novo FOXP1 Truncating Mutation in a Patient Originally Diagnosed as C Syndrome. Sci Rep 2018; 8:694. [PMID: 29330474 PMCID: PMC5766530 DOI: 10.1038/s41598-017-19109-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 12/21/2017] [Indexed: 01/04/2023] Open
Abstract
De novo FOXP1 mutations have been associated with intellectual disability (ID), motor delay, autistic features and a wide spectrum of speech difficulties. C syndrome (Opitz C trigonocephaly syndrome) is a rare and genetically heterogeneous condition, characterized by trigonocephaly, craniofacial anomalies and ID. Several different chromosome deletions and and point mutations in distinct genes have been associated with the disease in patients originally diagnosed as Opitz C. By whole exome sequencing we identified a de novo splicing mutation in FOXP1 in a patient, initially diagnosed as C syndrome, who suffers from syndromic intellectual disability with trigonocephaly. The mutation (c.1428 + 1 G > A) promotes the skipping of exon 16, a frameshift and a premature STOP codon (p.Ala450GLyfs*13), as assessed by a minigene strategy. The patient reported here shares speech difficulties, intellectual disability and autistic features with other FOXP1 syndrome patients, and thus the diagnosis for this patient should be changed. Finally, since trigonocephaly has not been previously reported in FOXP1 syndrome, it remains to be proved whether it may be associated with the FOXP1 mutation.
Collapse
Affiliation(s)
- Roser Urreizti
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, IBUB, IRSJD, CIBERER, Barcelona, Spain.
| | - Sarah Damanti
- Geriatric Unit, Fondazione Ca'Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy.,Nutritional Sciences, University of Milan, Milan, Italy
| | - Carla Esteve
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, IBUB, IRSJD, CIBERER, Barcelona, Spain
| | - Héctor Franco-Valls
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, IBUB, IRSJD, CIBERER, Barcelona, Spain
| | - Laura Castilla-Vallmanya
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, IBUB, IRSJD, CIBERER, Barcelona, Spain
| | - Raul Tonda
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Bru Cormand
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, IBUB, IRSJD, CIBERER, Barcelona, Spain
| | - Lluïsa Vilageliu
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, IBUB, IRSJD, CIBERER, Barcelona, Spain
| | - John M Opitz
- Pediatrics Medical Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Giovanni Neri
- Istituto di Medicina Genomica, Università Cattolica Sacro Cuore, Policlinico A. Gemelli, Rome, Italy
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, IBUB, IRSJD, CIBERER, Barcelona, Spain
| | - Susana Balcells
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, IBUB, IRSJD, CIBERER, Barcelona, Spain
| |
Collapse
|
65
|
Coffer P, Braccioli L, Nijboer C. Forkhead box protein P1, a key player in neuronal development? Neural Regen Res 2018; 13:801-802. [PMID: 29863003 PMCID: PMC5998630 DOI: 10.4103/1673-5374.232467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
66
|
Usui N, Araujo DJ, Kulkarni A, Co M, Ellegood J, Harper M, Toriumi K, Lerch JP, Konopka G. Foxp1 regulation of neonatal vocalizations via cortical development. Genes Dev 2017; 31:2039-2055. [PMID: 29138280 PMCID: PMC5733496 DOI: 10.1101/gad.305037.117] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/25/2017] [Indexed: 12/25/2022]
Abstract
Usui et al. show that deletion of Foxp1 in the developing forebrain leads to impairments in neonatal vocalizations as well as neocortical cytoarchitectonic alterations via neuronal positioning and migration. Sumoylation of Foxp1 affects neuronal differentiation and migration in the developing neocortex. The molecular mechanisms driving brain development at risk in autism spectrum disorders (ASDs) remain mostly unknown. Previous studies have implicated the transcription factor FOXP1 in both brain development and ASD pathophysiology. However, the specific molecular pathways both upstream of and downstream from FOXP1 are not fully understood. To elucidate the contribution of FOXP1-mediated signaling to brain development and, in particular, neocortical development, we generated forebrain-specific Foxp1 conditional knockout mice. We show that deletion of Foxp1 in the developing forebrain leads to impairments in neonatal vocalizations as well as neocortical cytoarchitectonic alterations via neuronal positioning and migration. Using a genomics approach, we identified the transcriptional networks regulated by Foxp1 in the developing neocortex and found that such networks are enriched for downstream targets involved in neurogenesis and neuronal migration. We also uncovered mechanistic insight into Foxp1 function by demonstrating that sumoylation of Foxp1 during embryonic brain development is necessary for mediating proper interactions between Foxp1 and the NuRD complex. Furthermore, we demonstrated that sumoylation of Foxp1 affects neuronal differentiation and migration in the developing neocortex. Together, these data provide critical mechanistic insights into the function of FOXP1 in the developing neocortex and may reveal molecular pathways at risk in ASD.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Division of Development of Mental Functions, Research Center for Child Mental Development, University of Fukui, Fukui 910-1193, Japan.,Division of Developmental Higher Brain Functions, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Osaka 565-0871, Japan
| | - Daniel J Araujo
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ashwinikumar Kulkarni
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Marissa Co
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Jacob Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Ontario M5S 1A1, Canada
| | - Matthew Harper
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Kazuya Toriumi
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Project for Schizophrenia Research, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Jason P Lerch
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Ontario M5S 1A1, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Genevieve Konopka
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
67
|
Siper PM, De Rubeis S, Trelles MDP, Durkin A, Di Marino D, Muratet F, Frank Y, Lozano R, Eichler EE, Kelly M, Beighley J, Gerdts J, Wallace AS, Mefford HC, Bernier RA, Kolevzon A, Buxbaum JD. Prospective investigation of FOXP1 syndrome. Mol Autism 2017; 8:57. [PMID: 29090079 PMCID: PMC5655854 DOI: 10.1186/s13229-017-0172-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
Background Haploinsufficiency of the forkhead-box protein P1 (FOXP1) gene leads to a neurodevelopmental disorder termed FOXP1 syndrome. Previous studies in individuals carrying FOXP1 mutations and deletions have described the presence of autism spectrum disorder (ASD) traits, intellectual disability, language impairment, and psychiatric features. The goal of the present study was to comprehensively characterize the genetic and clinical spectrum of FOXP1 syndrome. This is the first study to prospectively examine the genotype-phenotype relationship in multiple individuals with FOXP1 syndrome, using a battery of standardized clinical assessments. Methods Genetic and clinical data was obtained and analyzed from nine children and adolescents between the ages of 5–17 with mutations in FOXP1. Phenotypic characterization included gold standard ASD testing and norm-referenced measures of cognition, adaptive behavior, language, motor, and visual-motor integration skills. In addition, psychiatric, medical, neurological, and dysmorphology examinations were completed by a multidisciplinary team of clinicians. A comprehensive review of reported cases was also performed. All missense and in-frame mutations were mapped onto the three-dimensional structure of DNA-bound FOXP1. Results We have identified nine de novo mutations, including three frameshift, one nonsense, one mutation in an essential splice site resulting in frameshift and insertion of a premature stop codon, three missense, and one in-frame deletion. Reviewing prior literature, we found seven instances of recurrent mutations and another 34 private mutations. The majority of pathogenic missense and in-frame mutations, including all four missense mutations in our cohort, lie in the DNA-binding domain. Through structural analyses, we show that the mutations perturb amino acids necessary for binding to the DNA or interfere with the domain swapping that mediates FOXP1 dimerization. Individuals with FOXP1 syndrome presented with delays in early motor and language milestones, language impairment (expressive language > receptive language), ASD symptoms, visual-motor integration deficits, and complex psychiatric presentations characterized by anxiety, obsessive-compulsive traits, attention deficits, and externalizing symptoms. Medical features included non-specific structural brain abnormalities and dysmorphic features, endocrine and gastrointestinal problems, sleep disturbances, and sinopulmonary infections. Conclusions This study identifies novel FOXP1 mutations associated with FOXP1 syndrome, identifies recurrent mutations, and demonstrates significant clustering of missense mutations in the DNA-binding domain. Clinical findings confirm the role FOXP1 plays in development across multiple domains of functioning. The genetic findings can be incorporated into clinical genetics practice to improve accurate genetic diagnosis of FOXP1 syndrome and the clinical findings can inform monitoring and treatment of individuals with FOXP1 syndrome. Electronic supplementary material The online version of this article (10.1186/s13229-017-0172-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paige M Siper
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Silvia De Rubeis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Maria Del Pilar Trelles
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Allison Durkin
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Daniele Di Marino
- Department of Informatics, Institute of Computational Science, Università della Svizzera Italiana, Lugano, Switzerland
| | - François Muratet
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Yitzchak Frank
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Reymundo Lozano
- Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, Department of Psychiatry, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington, Seattle, WA USA
| | - Morgan Kelly
- Department of Psychiatry, University of Washington, Seattle, WA USA
| | | | - Jennifer Gerdts
- Department of Psychiatry, University of Washington, Seattle, WA USA
| | | | | | | | - Alexander Kolevzon
- Department of Psychiatry, Department of Pediatrics, Friedman Brain Institute, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Joseph D Buxbaum
- Department of Psychiatry, Department of Genetics and Genomic Sciences, Department of Neuroscience, Friedman Brain Institute, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
68
|
Foxp1 in Forebrain Pyramidal Neurons Controls Gene Expression Required for Spatial Learning and Synaptic Plasticity. J Neurosci 2017; 37:10917-10931. [PMID: 28978667 DOI: 10.1523/jneurosci.1005-17.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 09/12/2017] [Accepted: 09/14/2017] [Indexed: 12/28/2022] Open
Abstract
Genetic perturbations of the transcription factor Forkhead Box P1 (FOXP1) are causative for severe forms of autism spectrum disorder that are often comorbid with intellectual disability. Recent work has begun to reveal an important role for FoxP1 in brain development, but the brain-region-specific contributions of Foxp1 to autism and intellectual disability phenotypes have yet to be determined fully. Here, we describe Foxp1 conditional knock-out (Foxp1cKO) male and female mice with loss of Foxp1 in the pyramidal neurons of the neocortex and the CA1/CA2 subfields of the hippocampus. Foxp1cKO mice exhibit behavioral phenotypes that are of potential relevance to autism spectrum disorder, including hyperactivity, increased anxiety, communication impairments, and decreased sociability. In addition, Foxp1cKO mice have gross deficits in learning and memory tasks of relevance to intellectual disability. Using a genome-wide approach, we identified differentially expressed genes in the hippocampus of Foxp1cKO mice associated with synaptic function and development. Furthermore, using magnetic resonance imaging, we uncovered a significant reduction in the volumes of both the entire hippocampus as well as individual hippocampal subfields of Foxp1cKO mice. Finally, we observed reduced maintenance of LTP in area CA1 of the hippocampus in these mutant mice. Together, these data suggest that proper expression of Foxp1 in the pyramidal neurons of the forebrain is important for regulating gene expression pathways that contribute to specific behaviors reminiscent of those seen in autism and intellectual disability. In particular, Foxp1 regulation of gene expression appears to be crucial for normal hippocampal development, CA1 plasticity, and spatial learning.SIGNIFICANCE STATEMENT Loss-of-function mutations in the transcription factor Forkhead Box P1 (FOXP1) lead to autism spectrum disorder and intellectual disability. Understanding the potential brain-region-specific contributions of FOXP1 to disease-relevant phenotypes could be a critical first step in the management of patients with these mutations. Here, we report that Foxp1 conditional knock-out (Foxp1cKO) mice with loss of Foxp1 in the neocortex and hippocampus display autism and intellectual-disability-relevant behaviors. We also show that these phenotypes correlate with changes in both the genomic and physiological profiles of the hippocampus in Foxp1cKO mice. Our work demonstrates that brain-region-specific FOXP1 expression may relate to distinct, clinically relevant phenotypes.
Collapse
|
69
|
Sollis E, Deriziotis P, Saitsu H, Miyake N, Matsumoto N, Hoffer MJV, Ruivenkamp CAL, Alders M, Okamoto N, Bijlsma EK, Plomp AS, Fisher SE. Equivalent missense variant in the FOXP2 and FOXP1 transcription factors causes distinct neurodevelopmental disorders. Hum Mutat 2017; 38:1542-1554. [PMID: 28741757 DOI: 10.1002/humu.23303] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 07/11/2017] [Accepted: 07/22/2017] [Indexed: 12/28/2022]
Abstract
The closely related paralogues FOXP2 and FOXP1 encode transcription factors with shared functions in the development of many tissues, including the brain. However, while mutations in FOXP2 lead to a speech/language disorder characterized by childhood apraxia of speech (CAS), the clinical profile of FOXP1 variants includes a broader neurodevelopmental phenotype with global developmental delay, intellectual disability, and speech/language impairment. Using clinical whole-exome sequencing, we report an identical de novo missense FOXP1 variant identified in three unrelated patients. The variant, p.R514H, is located in the forkhead-box DNA-binding domain and is equivalent to the well-studied p.R553H FOXP2 variant that cosegregates with CAS in a large UK family. We present here for the first time a direct comparison of the molecular and clinical consequences of the same mutation affecting the equivalent residue in FOXP1 and FOXP2. Detailed functional characterization of the two variants in cell model systems revealed very similar molecular consequences, including aberrant subcellular localization, disruption of transcription factor activity, and deleterious effects on protein interactions. Nonetheless, clinical manifestations were broader and more severe in the three cases carrying the p.R514H FOXP1 variant than in individuals with the p.R553H variant related to CAS, highlighting divergent roles of FOXP2 and FOXP1 in neurodevelopment.
Collapse
Affiliation(s)
- Elliot Sollis
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Pelagia Deriziotis
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mariëtte J V Hoffer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Claudia A L Ruivenkamp
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariëlle Alders
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan
| | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Astrid S Plomp
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| |
Collapse
|
70
|
Reduced Expression of Foxp1 as a Contributing Factor in Huntington's Disease. J Neurosci 2017; 37:6575-6587. [PMID: 28550168 DOI: 10.1523/jneurosci.3612-16.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/08/2017] [Accepted: 05/11/2017] [Indexed: 12/31/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disease caused by a polyglutamine expansion in the huntington protein (htt). The neuropathological hallmark of HD is the loss of neurons in the striatum and, to a lesser extent, in the cortex. Foxp1 is a member of the Forkhead family of transcription factors expressed selectively in the striatum and the cortex. In the brain, three major Foxp1 isoforms are expressed: isoform-A (∼90 kDa), isoform-D (∼70 kDa), and isoform-C (∼50 kDa). We find that expression of Foxp1 isoform-A and -D is selectively reduced in the striatum and cortex of R6/2 HD mice as well as in the striatum of HD patients. Furthermore, expression of mutant htt in neurons results in the downregulation of Foxp1 Elevating expression of isoform-A or -D protects cortical neurons from death caused by the expression of mutant htt On the other hand, knockdown of Foxp1 promotes death in otherwise healthy neurons. Neuroprotection by Foxp1 is likely to be mediated by the transcriptional stimulation of the cell-cycle inhibitory protein p21Waf1/Cip1 Consistently, Foxp1 activates transcription of the p21Waf1/Cip1 gene promoter, and overexpression of Foxp1 in neurons results in the elevation of p21 expression. Moreover, knocking down of p21Waf1/Cip1 blocks the ability of Foxp1 to protect neurons from mut-Htt-induced neurotoxicity. We propose that the selective vulnerability of neurons of the striatum and cortex in HD is related to the loss of expression of Foxp1, a protein that is highly expressed in these neurons and required for their survival.SIGNIFICANCE STATEMENT Although the mutant huntingtin gene is expressed widely, neurons of the striatum and cortex are selectively affected in Huntington's disease (HD). Our results suggest that this selectivity is attributable to the reduced expression of Foxp1, a protein expressed selectively in striatal and cortical neurons that plays a neuroprotective role in these cells. We show that protection by Foxp1 involves stimulation of the p21Waf1/Cip1 (Cdkn1a) gene. Although three major Foxp1 isoforms (A, C, and D) are expressed in the brain, only isoform-A has been studied in the nervous system. We show that isoform-D is also expressed selectively, neuroprotective and downregulated in HD mice and patients. Our results suggest that Foxp1 might be an attractive therapeutic target for HD.
Collapse
|
71
|
Merchan-Sala P, Nardini D, Waclaw RR, Campbell K. Selective neuronal expression of the SoxE factor, Sox8, in direct pathway striatal projection neurons of the developing mouse brain. J Comp Neurol 2017; 525:2805-2819. [PMID: 28472858 DOI: 10.1002/cne.24232] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 01/24/2023]
Abstract
The striatum is the major component of the basal ganglia and is well known to play a key role in the control of motor function via balanced output from the indirect (iSPNs) and direct pathway striatal projection neurons (dSPNs). Little is known, however, about the molecular genetic mechanisms that control the formation of the iSPNs versus dSPNs. We show here that the SoxE family member, Sox8, is co-expressed with the dSPN markers, Isl1 and Ebf1, in the developing striatum. Moreover, dSPNs, as marked by Isl1-cre fate map, express Sox8 in the embryonic striatum and Sox8-EGFP BAC transgenic mice specifically reveal the direct pathway axons during development. These EGFP+ axons are first observed to reach their midbrain target, the substantia nigra pars reticulata (SNr), at E14 in the mouse with a robust connection observed already at birth. The selective expression of EGFP in dSPNs of Sox8-EGFP BAC mice is maintained at postnatal timepoints. Sox8 is known to be expressed in oligodendrocyte precursor cells (OPCs) together with other SoxE factors and we show here that the EGFP signal co-localizes with the OPC markers throughout the brain. Finally, we show that Sox8-EGFP BAC mice can be used to interrogate the altered dSPN development in Isl1 conditional mutants including aberrant axonal projections detected already at embryonic timepoints. Thus, Sox8 represents an early and specific marker of embryonic dSPNs and the Sox8-EGFP BAC transgenic mice are an excellent tool to study the development of basal ganglia circuitry.
Collapse
Affiliation(s)
- Paloma Merchan-Sala
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Diana Nardini
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ronald R Waclaw
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kenneth Campbell
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
72
|
Meta-analysis of GWAS of over 16,000 individuals with autism spectrum disorder highlights a novel locus at 10q24.32 and a significant overlap with schizophrenia. Mol Autism 2017; 8:21. [PMID: 28540026 PMCID: PMC5441062 DOI: 10.1186/s13229-017-0137-9] [Citation(s) in RCA: 355] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 04/05/2017] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Over the past decade genome-wide association studies (GWAS) have been applied to aid in the understanding of the biology of traits. The success of this approach is governed by the underlying effect sizes carried by the true risk variants and the corresponding statistical power to observe such effects given the study design and sample size under investigation. Previous ASD GWAS have identified genome-wide significant (GWS) risk loci; however, these studies were of only of low statistical power to identify GWS loci at the lower effect sizes (odds ratio (OR) <1.15). METHODS We conducted a large-scale coordinated international collaboration to combine independent genotyping data to improve the statistical power and aid in robust discovery of GWS loci. This study uses genome-wide genotyping data from a discovery sample (7387 ASD cases and 8567 controls) followed by meta-analysis of summary statistics from two replication sets (7783 ASD cases and 11359 controls; and 1369 ASD cases and 137308 controls). RESULTS We observe a GWS locus at 10q24.32 that overlaps several genes including PITX3, which encodes a transcription factor identified as playing a role in neuronal differentiation and CUEDC2 previously reported to be associated with social skills in an independent population cohort. We also observe overlap with regions previously implicated in schizophrenia which was further supported by a strong genetic correlation between these disorders (Rg = 0.23; P = 9 × 10-6). We further combined these Psychiatric Genomics Consortium (PGC) ASD GWAS data with the recent PGC schizophrenia GWAS to identify additional regions which may be important in a common neurodevelopmental phenotype and identified 12 novel GWS loci. These include loci previously implicated in ASD such as FOXP1 at 3p13, ATP2B2 at 3p25.3, and a 'neurodevelopmental hub' on chromosome 8p11.23. CONCLUSIONS This study is an important step in the ongoing endeavour to identify the loci which underpin the common variant signal in ASD. In addition to novel GWS loci, we have identified a significant genetic correlation with schizophrenia and association of ASD with several neurodevelopmental-related genes such as EXT1, ASTN2, MACROD2, and HDAC4.
Collapse
|
73
|
Lee E, Lee J, Kim E. Excitation/Inhibition Imbalance in Animal Models of Autism Spectrum Disorders. Biol Psychiatry 2017; 81:838-847. [PMID: 27450033 DOI: 10.1016/j.biopsych.2016.05.011] [Citation(s) in RCA: 315] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/02/2016] [Accepted: 05/16/2016] [Indexed: 12/19/2022]
Abstract
Imbalances between excitation and inhibition in synaptic transmission and neural circuits have been implicated in autism spectrum disorders. Excitation and inhibition imbalances are frequently observed in animal models of autism spectrum disorders, and their correction normalizes key autistic-like phenotypes in these animals. These results suggest that excitation and inhibition imbalances may contribute to the development and maintenance of autism spectrum disorders and represent an important therapeutic target.
Collapse
Affiliation(s)
- Eunee Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Korea
| | - Jiseok Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea.
| |
Collapse
|
74
|
Fröhlich H, Rafiullah R, Schmitt N, Abele S, Rappold GA. Foxp1 expression is essential for sex-specific murine neonatal ultrasonic vocalization. Hum Mol Genet 2017; 26:1511-1521. [PMID: 28204507 DOI: 10.1093/hmg/ddx055] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/07/2017] [Indexed: 11/14/2022] Open
Abstract
Autism and speech and language deficits are predominantly found in boys, however the causative mechanisms for this sex bias are unknown. Human FOXP1 is associated with autism, intellectual disability and speech and language deficits. Its closely related family member FOXP2 is involved in speech and language disorder and Foxp2 deficient mice have demonstrated an absence of ultrasonic vocalizations (USVs). Since Foxp1 and Foxp2 form heterodimers for transcriptional regulation, we investigated USV in neonatal brain-specific Foxp1 KO mice. Foxp1 KO pups had strongly reduced USV and lacked the sex-specific call rate from WT pups, indicating that Foxp1 is essential for normal USV. As expression differences of Foxp1 or Foxp2 could explain the sex-dimorphic vocalization in WT animals, we quantified both proteins in the striatum and cortex at P7.5 and detected a sex-specific expression of Foxp2 in the striatum. We further analyzed Foxp1 and Foxp2 expression in the striatum and cortex of CD1 mice at different embryonic and postnatal stages and observed sex differences in both genes at E17.5 and P7.5. Sex hormones, especially androgens are known to play a crucial role in the sexual differentiation of vocalizations in many vertebrates. We show that Foxp1 and the androgen receptor are co-expressed in striatal medium spiny neurons and that brain-specific androgen receptor KO (ArNesCre) mice exhibit reduced Foxp1 expression in the striatum at E17.5 and P7.5 and an increased Foxp2 level in the cortex at P7.5. Thus, androgens may contribute to sex-specific differences in Foxp1 and Foxp2 expression and USV.
Collapse
Affiliation(s)
- Henning Fröhlich
- Department of Human Molecular Genetics, Medical Faculty of the Heidelberg University, Im Neuenheimer Feld 366, Heidelberg, Germany
| | - Rafiullah Rafiullah
- Department of Human Molecular Genetics, Medical Faculty of the Heidelberg University, Im Neuenheimer Feld 366, Heidelberg, Germany
| | - Nathalie Schmitt
- Department of Human Molecular Genetics, Medical Faculty of the Heidelberg University, Im Neuenheimer Feld 366, Heidelberg, Germany
| | - Sonja Abele
- Department of Human Molecular Genetics, Medical Faculty of the Heidelberg University, Im Neuenheimer Feld 366, Heidelberg, Germany
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, Medical Faculty of the Heidelberg University, Im Neuenheimer Feld 366, Heidelberg, Germany.,Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
75
|
Le Hellard S, Wang Y, Witoelar A, Zuber V, Bettella F, Hugdahl K, Espeseth T, Steen VM, Melle I, Desikan R, Schork AJ, Thompson WK, Dale AM, Djurovic S, Andreassen OA. Identification of Gene Loci That Overlap Between Schizophrenia and Educational Attainment. Schizophr Bull 2017; 43:654-664. [PMID: 27338279 PMCID: PMC5463752 DOI: 10.1093/schbul/sbw085] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
There is evidence for genetic overlap between cognitive abilities and schizophrenia (SCZ), and genome-wide association studies (GWAS) demonstrate that both SCZ and general cognitive abilities have a strong polygenic component with many single-nucleotide polymorphisms (SNPs) each with a small effect. Here we investigated the shared genetic architecture between SCZ and educational attainment, which is regarded as a "proxy phenotype" for cognitive abilities, but may also reflect other traits. We applied a conditional false discovery rate (condFDR) method to GWAS of SCZ (n = 82 315), college completion ("College," n = 95 427), and years of education ("EduYears," n = 101 069). Variants associated with College or EduYears showed enrichment of association with SCZ, demonstrating polygenic overlap. This was confirmed by an increased replication rate in SCZ. By applying a condFDR threshold <0.01, we identified 18 genomic loci associated with SCZ after conditioning on College and 15 loci associated with SCZ after conditioning on EduYears. Ten of these loci overlapped. Using conjunctional FDR, we identified 10 loci shared between SCZ and College, and 29 loci shared between SCZ and EduYears. The majority of these loci had effects in opposite directions. Our results provide evidence for polygenic overlap between SCZ and educational attainment, and identify novel pleiotropic loci. Other studies have reported genetic overlap between SCZ and cognition, or SCZ and educational attainment, with negative correlation. Importantly, our methods enable identification of bi-directional effects, which highlight the complex relationship between SCZ and educational attainment, and support polygenic mechanisms underlying both cognitive dysfunction and creativity in SCZ.
Collapse
Affiliation(s)
- Stéphanie Le Hellard
- NORMENT—KG Jebsen Centre, Department of Clinical Science, University of Bergen, Bergen, Norway;,Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Yunpeng Wang
- NORMENT—KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway;,NORMENT—KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway;,Multimodal Imaging Lab, University of California at San Diego, La Jolla, CA;,Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Aree Witoelar
- NORMENT—KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway;,NORMENT—KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Verena Zuber
- NORMENT—KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway;,NORMENT—KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway;,NCMM, University of Oslo, Oslo, Norway
| | - Francesco Bettella
- NORMENT—KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway;,NORMENT—KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Kenneth Hugdahl
- NORMENT, Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway;,Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Thomas Espeseth
- NORMENT—KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway;,Department of Psychology, University of Oslo, Oslo, Norway
| | - Vidar M. Steen
- NORMENT—KG Jebsen Centre, Department of Clinical Science, University of Bergen, Bergen, Norway;,Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Ingrid Melle
- NORMENT—KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway;,NORMENT—KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Rahul Desikan
- Multimodal Imaging Lab, University of California at San Diego, La Jolla, CA;,Department of Radiology, University of California at San Diego, La Jolla, CA
| | - Andrew J. Schork
- Multimodal Imaging Lab, University of California at San Diego, La Jolla, CA;,Department of Cognitive Science, University of California San Diego, La Jolla, CA
| | - Wesley K. Thompson
- Department of Psychiatry, University of California, San Diego, La Jolla, CA
| | - Anders M. Dale
- Multimodal Imaging Lab, University of California at San Diego, La Jolla, CA;,Department of Cognitive Science, University of California San Diego, La Jolla, CA;,Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Srdjan Djurovic
- NORMENT—KG Jebsen Centre, Department of Clinical Science, University of Bergen, Bergen, Norway;,NORMENT—KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway;,Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Ole A. Andreassen
- NORMENT—KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway;,NORMENT—KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway;,Multimodal Imaging Lab, University of California at San Diego, La Jolla, CA
| | | |
Collapse
|
76
|
Rocca DL, Wilkinson KA, Henley JM. SUMOylation of FOXP1 regulates transcriptional repression via CtBP1 to drive dendritic morphogenesis. Sci Rep 2017; 7:877. [PMID: 28408745 PMCID: PMC5429823 DOI: 10.1038/s41598-017-00707-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/08/2017] [Indexed: 12/13/2022] Open
Abstract
Forkhead Box P (FOXP) transcriptional repressors play a major role in brain development and their dysfunction leads to human cognitive disorders. However, little is known about how the activity of these proteins is regulated. Here, we show that FOXP1 SUMOylation at lysine 670 is required for recruiting the co-repressor CtBP1 and transcriptional repression. FOXP1 SUMOylation is tightly controlled by neuronal activity, in which synapse to nucleus signalling, mediated via NMDAR and L-type calcium channels, results in rapid FOXP1 deSUMOylation. Knockdown of FOXP1 in cultured cortical neurons stunts dendritic outgrowth and this phenotype cannot be rescued by replacement with a non-SUMOylatable FOXP1-K670R mutant, indicating that SUMOylation of FOXP1 is essential for regulation of proper neuronal morphogenesis. These results suggest that activity-dependent SUMOylation of FOXP1 may be an important mediator of early cortical development and neuronal network formation in the brain.
Collapse
Affiliation(s)
- Daniel L Rocca
- School of Biochemistry, Centre for Synaptic Plasticity, Medical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Kevin A Wilkinson
- School of Biochemistry, Centre for Synaptic Plasticity, Medical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Jeremy M Henley
- School of Biochemistry, Centre for Synaptic Plasticity, Medical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
77
|
Chen R, Davis LK, Guter S, Wei Q, Jacob S, Potter MH, Cox NJ, Cook EH, Sutcliffe JS, Li B. Leveraging blood serotonin as an endophenotype to identify de novo and rare variants involved in autism. Mol Autism 2017; 8:14. [PMID: 28344757 PMCID: PMC5361831 DOI: 10.1186/s13229-017-0130-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 03/10/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is one of the most highly heritable neuropsychiatric disorders, but underlying molecular mechanisms are still unresolved due to extreme locus heterogeneity. Leveraging meaningful endophenotypes or biomarkers may be an effective strategy to reduce heterogeneity to identify novel ASD genes. Numerous lines of evidence suggest a link between hyperserotonemia, i.e., elevated serotonin (5-hydroxytryptamine or 5-HT) in whole blood, and ASD. However, the genetic determinants of blood 5-HT level and their relationship to ASD are largely unknown. METHODS In this study, pursuing the hypothesis that de novo variants (DNVs) and rare risk alleles acting in a recessive mode may play an important role in predisposition of hyperserotonemia in people with ASD, we carried out whole exome sequencing (WES) in 116 ASD parent-proband trios with most (107) probands having 5-HT measurements. RESULTS Combined with published ASD DNVs, we identified USP15 as having recurrent de novo loss of function mutations and discovered evidence supporting two other known genes with recurrent DNVs (FOXP1 and KDM5B). Genes harboring functional DNVs significantly overlap with functional/disease gene sets known to be involved in ASD etiology, including FMRP targets and synaptic formation and transcriptional regulation genes. We grouped the probands into High-5HT and Normal-5HT groups based on normalized serotonin levels, and used network-based gene set enrichment analysis (NGSEA) to identify novel hyperserotonemia-related ASD genes based on LoF and missense DNVs. We found enrichment in the High-5HT group for a gene network module (DAWN-1) previously implicated in ASD, and this points to the TGF-β pathway and cell junction processes. Through analysis of rare recessively acting variants (RAVs), we also found that rare compound heterozygotes (CHs) in the High-5HT group were enriched for loci in an ASD-associated gene set. Finally, we carried out rare variant group-wise transmission disequilibrium tests (gTDT) and observed significant association of rare variants in genes encoding a subset of the serotonin pathway with ASD. CONCLUSIONS Our study identified USP15 as a novel gene implicated in ASD based on recurrent DNVs. It also demonstrates the potential value of 5-HT as an effective endophenotype for gene discovery in ASD, and the effectiveness of this strategy needs to be further explored in studies of larger sample sizes.
Collapse
Affiliation(s)
- Rui Chen
- Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN USA.,Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA
| | - Lea K Davis
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA.,Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN USA
| | - Stephen Guter
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL USA
| | - Qiang Wei
- Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN USA.,Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA
| | - Suma Jacob
- Department of Psychiatry, University of Minnesota, Minneapolis, MN USA
| | - Melissa H Potter
- Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN USA.,Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA
| | - Nancy J Cox
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA.,Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN USA
| | - Edwin H Cook
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL USA
| | - James S Sutcliffe
- Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN USA.,Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA
| | - Bingshan Li
- Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN USA.,Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA
| |
Collapse
|
78
|
Peter S, De Zeeuw CI, Boeckers TM, Schmeisser MJ. Cerebellar and Striatal Pathologies in Mouse Models of Autism Spectrum Disorder. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2017; 224:103-119. [PMID: 28551753 DOI: 10.1007/978-3-319-52498-6_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition with a strong genetic component. To date, several hundred different genetic mutations have been identified to play a role in its aetiology. The heterogeneity of genetic abnormalities combined with the different brain regions where aberrations are found makes the search for causative mechanisms a daunting task. Even within a limited number of brain regions, a myriad of different neural circuit dysfunctions may lead to ASD. Here, we review mouse models that incorporate mutations of ASD risk genes causing pathologies in the cerebellum and striatum and highlight the vulnerability of related circuit dysfunctions within these brain regions in ASD pathophysiology.
Collapse
Affiliation(s)
- Saša Peter
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands. .,Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Chris I De Zeeuw
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.,Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Michael J Schmeisser
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany. .,Division of Neuroanatomy, Institute of Anatomy, Otto-von-Guericke University, Magdeburg, Germany. .,Leibniz Institute for Neurobiology, Magdeburg, Germany.
| |
Collapse
|
79
|
Abstract
Forkhead box (Fox) transcription factors are evolutionarily conserved in organisms ranging from yeast to humans. They regulate diverse biological processes both during development and throughout adult life. Mutations in many Fox genes are associated with human disease and, as such, various animal models have been generated to study the function of these transcription factors in mechanistic detail. In many cases, the absence of even a single Fox transcription factor is lethal. In this Primer, we provide an overview of the Fox family, highlighting several key Fox transcription factor families that are important for mammalian development.
Collapse
Affiliation(s)
- Maria L Golson
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
80
|
Atsem S, Reichenbach J, Potabattula R, Dittrich M, Nava C, Depienne C, Böhm L, Rost S, Hahn T, Schorsch M, Haaf T, El Hajj N. Paternal age effects on sperm FOXK1 and KCNA7 methylation and transmission into the next generation. Hum Mol Genet 2016; 25:4996-5005. [PMID: 28171595 PMCID: PMC5418740 DOI: 10.1093/hmg/ddw328] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/16/2016] [Accepted: 09/21/2016] [Indexed: 01/27/2023] Open
Abstract
Children of older fathers carry an increased risk for developing autism and other disorders. To elucidate the underlying mechanisms, we investigated the correlation of sperm DNA methylation with paternal age and its impact on the epigenome of the offspring. Methylation levels of nine candidate genes and LINE-1 repeats were quantified by bisulfite pyrosequencing in sperm DNA of 162 donors and 191 cord blood samples of resulting children (conceived by IVF/ICSI with the same sperm samples). Four genes showed a significant negative correlation between sperm methylation and paternal age. For FOXK1 and KCNA7, the age effect on the sperm epigenome was replicated in an independent cohort of 188 sperm samples. For FOXK1, paternal age also significantly correlated with foetal cord blood (FCB) methylation. Deep bisulfite sequencing and allele-specific pyrosequencing allowed us to distinguish between maternal and paternal alleles in FCB samples with an informative SNP. FCB methylation of the paternal FOXK1 allele was negatively correlated with paternal age, whereas maternal allele was unaffected by maternal age. Since FOXK1 duplication has been associated with autism, we studied blood FOXK1 methylation in 74 children with autism and 41 age-matched controls. The FOXK1 promoter showed a trend for accelerated demethylation in the autism group. Dual luciferase reporter assay revealed that FOXK1 methylation influences gene expression. Collectively, our study demonstrates that age-related DNA methylation changes in sperm can be transmitted to the next generation and may contribute to the increased disease risk in offspring of older fathers.
Collapse
Affiliation(s)
- Stefanie Atsem
- Institute of Human Genetics, Julius-Maximilians University, Würzburg, Germany
| | - Juliane Reichenbach
- Institute of Human Genetics, Julius-Maximilians University, Würzburg, Germany
| | - Ramya Potabattula
- Institute of Human Genetics, Julius-Maximilians University, Würzburg, Germany
| | - Marcus Dittrich
- Institute of Human Genetics, Julius-Maximilians University, Würzburg, Germany
| | - Caroline Nava
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Christel Depienne
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
- Département de Médicine translationnelle et Neurogénétique, IGBMC, CNRS UMR 7104/INSERM U964/Université de Strasbourg, Illkirch, France
| | - Lena Böhm
- Institute of Human Genetics, Julius-Maximilians University, Würzburg, Germany
| | - Simone Rost
- Institute of Human Genetics, Julius-Maximilians University, Würzburg, Germany
| | | | | | - Thomas Haaf
- Institute of Human Genetics, Julius-Maximilians University, Würzburg, Germany
| | - Nady El Hajj
- Institute of Human Genetics, Julius-Maximilians University, Würzburg, Germany
| |
Collapse
|
81
|
Yin J, Schaaf CP. Autism genetics - an overview. Prenat Diagn 2016; 37:14-30. [DOI: 10.1002/pd.4942] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/04/2016] [Accepted: 10/11/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Jiani Yin
- Department of Molecular and Human Genetics; Baylor College of Medicine; Houston TX USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital; Houston TX USA
| | - Christian P. Schaaf
- Department of Molecular and Human Genetics; Baylor College of Medicine; Houston TX USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital; Houston TX USA
| |
Collapse
|
82
|
Bakshi A, Zhu Z, Vinkhuyzen AAE, Hill WD, McRae AF, Visscher PM, Yang J. Fast set-based association analysis using summary data from GWAS identifies novel gene loci for human complex traits. Sci Rep 2016; 6:32894. [PMID: 27604177 PMCID: PMC5015118 DOI: 10.1038/srep32894] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/17/2016] [Indexed: 12/21/2022] Open
Abstract
We propose a method (fastBAT) that performs a fast set-based association analysis for human complex traits using summary-level data from genome-wide association studies (GWAS) and linkage disequilibrium (LD) data from a reference sample with individual-level genotypes. We demonstrate using simulations and analyses of real datasets that fastBAT is more accurate and orders of magnitude faster than the prevailing methods. Using fastBAT, we analyze summary data from the latest meta-analyses of GWAS on 150,064-339,224 individuals for height, body mass index (BMI), and schizophrenia. We identify 6 novel gene loci for height, 2 for BMI, and 3 for schizophrenia at PfastBAT < 5 × 10(-8). The gain of power is due to multiple small independent association signals at these loci (e.g. the THRB and FOXP1 loci for schizophrenia). The method is general and can be applied to GWAS data for all complex traits and diseases in humans and to such data in other species.
Collapse
Affiliation(s)
- Andrew Bakshi
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
- Centre for Systems Genomics, School of BioSciences, The University of Melbourne, Parkville 3010, Victoria, Australia
| | - Zhihong Zhu
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Anna A. E. Vinkhuyzen
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - W. David Hill
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, 7 George Square, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Allan F. McRae
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Peter M. Visscher
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- The University of Queensland Diamantina Institute, The Translation Research Institute, Brisbane, Queensland, Australia
| | - Jian Yang
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- The University of Queensland Diamantina Institute, The Translation Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
83
|
Srinivasan S, Bettella F, Mattingsdal M, Wang Y, Witoelar A, Schork AJ, Thompson WK, Zuber V, Winsvold BS, Zwart JA, Collier DA, Desikan RS, Melle I, Werge T, Dale AM, Djurovic S, Andreassen OA. Genetic Markers of Human Evolution Are Enriched in Schizophrenia. Biol Psychiatry 2016; 80:284-292. [PMID: 26681495 PMCID: PMC5397584 DOI: 10.1016/j.biopsych.2015.10.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 09/30/2015] [Accepted: 10/05/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Why schizophrenia has accompanied humans throughout our history despite its negative effect on fitness remains an evolutionary enigma. It is proposed that schizophrenia is a by-product of the complex evolution of the human brain and a compromise for humans' language, creative thinking, and cognitive abilities. METHODS We analyzed recent large genome-wide association studies of schizophrenia and a range of other human phenotypes (anthropometric measures, cardiovascular disease risk factors, immune-mediated diseases) using a statistical framework that draws on polygenic architecture and ancillary information on genetic variants. We used information from the evolutionary proxy measure called the Neanderthal selective sweep (NSS) score. RESULTS Gene loci associated with schizophrenia are significantly (p = 7.30 × 10(-9)) more prevalent in genomic regions that are likely to have undergone recent positive selection in humans (i.e., with a low NSS score). Variants in brain-related genes with a low NSS score confer significantly higher susceptibility than variants in other brain-related genes. The enrichment is strongest for schizophrenia, but we cannot rule out enrichment for other phenotypes. The false discovery rate conditional on the evolutionary proxy points to 27 candidate schizophrenia susceptibility loci, 12 of which are associated with schizophrenia and other psychiatric disorders or linked to brain development. CONCLUSIONS Our results suggest that there is a polygenic overlap between schizophrenia and NSS score, a marker of human evolution, which is in line with the hypothesis that the persistence of schizophrenia is related to the evolutionary process of becoming human.
Collapse
Affiliation(s)
- Saurabh Srinivasan
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Francesco Bettella
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Morten Mattingsdal
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Yunpeng Wang
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Multimodal Imaging Laboratory, University of California at San Diego, La Jolla, CA, USA
| | - Aree Witoelar
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Andrew J. Schork
- Multimodal Imaging Laboratory, University of California at San Diego, La Jolla, CA, USA,Cognitive Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA,Center for Human Development, University of California at San Diego, La Jolla, CA, USA
| | - Wesley K. Thompson
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Verena Zuber
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | | | - Bendik S. Winsvold
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology and FORMI, Oslo University Hospital, Ullevål, Oslo, Norway
| | - John-Anker Zwart
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology and FORMI, Oslo University Hospital, Ullevål, Oslo, Norway
| | | | - Rahul S. Desikan
- Multimodal Imaging Laboratory, University of California at San Diego, La Jolla, CA, USA,Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, CA, USA
| | - Ingrid Melle
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Center St. Hans, Mental Health Services Copenhagen, Roskilde, Denmark. Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark. The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark
| | - Anders M. Dale
- Multimodal Imaging Laboratory, University of California at San Diego, La Jolla, CA, USA,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,Lilly UK, Erl Wood Manor, Windlesham, Surrey, UK,Department of Neuroscience, University of California at San Diego, La Jolla, CA, USA
| | - Srdjan Djurovic
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ole A. Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,Corresponding author: , NORMENT, KG Jebsen Centre for Psychosis Research, Oslo University Hospital - Ullevål, Kirkeveien 166, PO Box 4956 Nydalen, 0424 Oslo, Norway; Tel.: +47- 22 11 98 90, Fax: +47- 22 11 98 99
| |
Collapse
|
84
|
Konopka G, Roberts TF. Insights into the Neural and Genetic Basis of Vocal Communication. Cell 2016; 164:1269-1276. [PMID: 26967292 DOI: 10.1016/j.cell.2016.02.039] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Indexed: 12/11/2022]
Abstract
The use of vocalizations to communicate information and elaborate social bonds is an adaptation seen in many vertebrate species. Human speech is an extreme version of this pervasive form of communication. Unlike the vocalizations exhibited by the majority of land vertebrates, speech is a learned behavior requiring early sensory exposure and auditory feedback for its development and maintenance. Studies in humans and a small number of other species have provided insights into the neural and genetic basis for learned vocal communication and are helping to delineate the roles of brain circuits across the cortex, basal ganglia, and cerebellum in generating vocal behaviors. This Review provides an outline of the current knowledge about these circuits and the genes implicated in vocal communication, as well as a perspective on future research directions in this field.
Collapse
Affiliation(s)
- Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Todd F Roberts
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| |
Collapse
|
85
|
Perinatal reduction of functional serotonin transporters results in developmental delay. Neuropharmacology 2016; 109:96-111. [PMID: 27208789 DOI: 10.1016/j.neuropharm.2016.05.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 03/25/2016] [Accepted: 05/17/2016] [Indexed: 11/22/2022]
Abstract
While there is strong evidence from rodent and human studies that a reduction in serotonin transporter (5-HTT) function in early-life can increase the risk for several neuropsychiatric disorders in adulthood, the effects of reduced 5-HTT function on behavior across developmental stages are underinvestigated. To elucidate how perinatal pharmacological and lifelong genetic inactivation of the 5-HTT affects behavior across development, we conducted a battery of behavioral tests in rats perinatally exposed to fluoxetine or vehicle and in 5-HTT(-/-) versus 5-HTT(+/+) rats. We measured motor-related behavior, olfactory function, grooming behavior, sensorimotor gating, object directed behavior and novel object recognition in the first three postnatal weeks and if possible the tests were repeated in adolescence and adulthood. We also measured developmental milestones such as eye opening, reflex development and body weight. We observed that both pharmacological and genetic inactivation of 5-HTT resulted in a developmental delay. Except for hypo-locomotion, most of the observed early-life effects were normalized later in life. In adolescence and adulthood we observed object directed behavior and decreased novel object recognition in the 5-HTT(-/-) rats, which might be related to the lifelong inactivation of 5-HTT. Together, these data provide an important contribution to the understanding of the effects of perinatal and lifelong 5-HTT inactivation on behavior across developmental stages.
Collapse
|
86
|
Altieri SC, Zhao T, Jalabi W, Romito-DiGiacomo RR, Maricich SM. En1 is necessary for survival of neurons in the ventral nuclei of the lateral lemniscus. Dev Neurobiol 2016; 76:1266-1274. [PMID: 26914477 DOI: 10.1002/dneu.22388] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 02/03/2016] [Accepted: 02/18/2016] [Indexed: 11/06/2022]
Abstract
The ventral nuclei of the lateral lemniscus (VNLL) are part of the central auditory system thought to participate in temporal sound processing. While the timing and location of VNLL neurogenesis have been determined, the genetic factors that regulate VNLL neuron development are unknown. Here, we use genetic fate-mapping techniques to demonstrate that all glycinergic and glycinergic/GABAergic VNLL neurons derive from a cellular lineage that expresses the homeobox transcription factor Engrailed 1 (En1). We also show that En1 deletion does not affect migration or adoption of a neuronal cell fate but does lead to VNLL neuron death during development. Furthermore, En1 deletion blocks expression of the transcription factor FoxP1 in a subset of VNLL neurons. Together, these data identify En1 as a gene important for VNLL neuron development and survival. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1266-1274, 2016.
Collapse
Affiliation(s)
- Stefanie C Altieri
- Richard King Mellon Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, 15224
| | - Tianna Zhao
- Richard King Mellon Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, 15224
| | - Walid Jalabi
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, 44106
| | | | - Stephen M Maricich
- Richard King Mellon Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, 15224. .,Childrens' Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, 15224.
| |
Collapse
|
87
|
Araujo DJ, Anderson AG, Berto S, Runnels W, Harper M, Ammanuel S, Rieger MA, Huang HC, Rajkovich K, Loerwald KW, Dekker JD, Tucker HO, Dougherty JD, Gibson JR, Konopka G. FoxP1 orchestration of ASD-relevant signaling pathways in the striatum. Genes Dev 2016; 29:2081-96. [PMID: 26494785 PMCID: PMC4617974 DOI: 10.1101/gad.267989.115] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In this study, Araujo et al. demonstrate that Foxp1 plays a role in the transcriptional regulation of autism-related pathways as well as genes involved in neuronal activity by identifying the gene expression program regulated by FoxP1 in both human neural cells and patient-relevant heterozygous Foxp1 mouse brains. Mutations in the transcription factor Forkhead box p1 (FOXP1) are causative for neurodevelopmental disorders such as autism. However, the function of FOXP1 within the brain remains largely uncharacterized. Here, we identify the gene expression program regulated by FoxP1 in both human neural cells and patient-relevant heterozygous Foxp1 mouse brains. We demonstrate a role for FoxP1 in the transcriptional regulation of autism-related pathways as well as genes involved in neuronal activity. We show that Foxp1 regulates the excitability of striatal medium spiny neurons and that reduction of Foxp1 correlates with defects in ultrasonic vocalizations. Finally, we demonstrate that FoxP1 has an evolutionarily conserved role in regulating pathways involved in striatal neuron identity through gene expression studies in human neural progenitors with altered FOXP1 levels. These data support an integral role for FoxP1 in regulating signaling pathways vulnerable in autism and the specific regulation of striatal pathways important for vocal communication.
Collapse
Affiliation(s)
- Daniel J Araujo
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ashley G Anderson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Stefano Berto
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Wesley Runnels
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Matthew Harper
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Simon Ammanuel
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Michael A Rieger
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Hung-Chung Huang
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Department of Biology, Jackson State University, Jackson, Mississippi 39217, USA
| | - Kacey Rajkovich
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Kristofer W Loerwald
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Joseph D Dekker
- University of Texas at Austin, Section of Molecular Genetics and Microbiology, Austin, Texas 78712, USA
| | - Haley O Tucker
- University of Texas at Austin, Section of Molecular Genetics and Microbiology, Austin, Texas 78712, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Jay R Gibson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Genevieve Konopka
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
88
|
Kochinke K, Zweier C, Nijhof B, Fenckova M, Cizek P, Honti F, Keerthikumar S, Oortveld M, Kleefstra T, Kramer J, Webber C, Huynen M, Schenck A. Systematic Phenomics Analysis Deconvolutes Genes Mutated in Intellectual Disability into Biologically Coherent Modules. Am J Hum Genet 2016; 98:149-64. [PMID: 26748517 DOI: 10.1016/j.ajhg.2015.11.024] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/25/2015] [Indexed: 10/22/2022] Open
Abstract
Intellectual disability (ID) disorders are genetically and phenotypically extremely heterogeneous. Can this complexity be depicted in a comprehensive way as a means of facilitating the understanding of ID disorders and their underlying biology? We provide a curated database of 746 currently known genes, mutations in which cause ID (ID-associated genes [ID-AGs]), classified according to ID manifestation and associated clinical features. Using this integrated resource, we show that ID-AGs are substantially enriched with co-expression, protein-protein interactions, and specific biological functions. Systematic identification of highly enriched functional themes and phenotypes revealed typical phenotype combinations characterizing process-defined groups of ID disorders, such as chromatin-related disorders and deficiencies in DNA repair. Strikingly, phenotype classification efficiently breaks down ID-AGs into subsets with significantly elevated biological coherence and predictive power. Custom-made functional Drosophila datasets revealed further characteristic phenotypes among ID-AGs and specific clinical classes. Our study and resource provide systematic insights into the molecular and clinical landscape of ID disorders, represent a significant step toward overcoming current limitations in ID research, and prove the utility of systematic human and cross-species phenomics analyses in highly heterogeneous genetic disorders.
Collapse
|
89
|
Sollis E, Graham SA, Vino A, Froehlich H, Vreeburg M, Dimitropoulou D, Gilissen C, Pfundt R, Rappold GA, Brunner HG, Deriziotis P, Fisher SE. Identification and functional characterization of de novo FOXP1 variants provides novel insights into the etiology of neurodevelopmental disorder. Hum Mol Genet 2015; 25:546-57. [PMID: 26647308 DOI: 10.1093/hmg/ddv495] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/27/2015] [Indexed: 12/27/2022] Open
Abstract
De novo disruptions of the neural transcription factor FOXP1 are a recently discovered, rare cause of sporadic intellectual disability (ID). We report three new cases of FOXP1-related disorder identified through clinical whole-exome sequencing. Detailed phenotypic assessment confirmed that global developmental delay, autistic features, speech/language deficits, hypotonia and mild dysmorphic features are core features of the disorder. We expand the phenotypic spectrum to include sensory integration disorder and hypertelorism. Notably, the etiological variants in these cases include two missense variants within the DNA-binding domain of FOXP1. Only one such variant has been reported previously. The third patient carries a stop-gain variant. We performed functional characterization of the three missense variants alongside our stop-gain and two previously described truncating/frameshift variants. All variants severely disrupted multiple aspects of protein function. Strikingly, the missense variants had similarly severe effects on protein function as the truncating/frameshift variants. Our findings indicate that a loss of transcriptional repression activity of FOXP1 underlies the neurodevelopmental phenotype in FOXP1-related disorder. Interestingly, the three novel variants retained the ability to interact with wild-type FOXP1, suggesting these variants could exert a dominant-negative effect by interfering with the normal FOXP1 protein. These variants also retained the ability to interact with FOXP2, a paralogous transcription factor disrupted in rare cases of speech and language disorder. Thus, speech/language deficits in these individuals might be worsened through deleterious effects on FOXP2 function. Our findings highlight that de novo FOXP1 variants are a cause of sporadic ID and emphasize the importance of this transcription factor in neurodevelopment.
Collapse
Affiliation(s)
- Elliot Sollis
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen 6525 XD, The Netherlands
| | - Sarah A Graham
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen 6525 XD, The Netherlands
| | - Arianna Vino
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen 6525 XD, The Netherlands
| | - Henning Froehlich
- Department of Human Molecular Genetics, Heidelberg University, Heidelberg 69120, Germany
| | - Maaike Vreeburg
- Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW), Maastricht UMC, Maastricht 6202 AZ, The Netherlands
| | - Danai Dimitropoulou
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen 6525 XD, The Netherlands
| | - Christian Gilissen
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Nijmegen 6500 HB, The Netherlands
| | - Rolph Pfundt
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Nijmegen 6500 HB, The Netherlands
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, Heidelberg University, Heidelberg 69120, Germany, Interdisciplinary Center of Neurosciences (IZN), Heidelberg University, Heidelberg 69120, Germany and
| | - Han G Brunner
- Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW), Maastricht UMC, Maastricht 6202 AZ, The Netherlands, Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Nijmegen 6500 HB, The Netherlands
| | - Pelagia Deriziotis
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen 6525 XD, The Netherlands,
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen 6525 XD, The Netherlands, Donders Institute for Brain, Cognition and Behaviour, Nijmegen 6525 EN, The Netherlands
| |
Collapse
|
90
|
Golonzhka O, Nord A, Tang PLF, Lindtner S, Ypsilanti AR, Ferretti E, Visel A, Selleri L, Rubenstein JLR. Pbx Regulates Patterning of the Cerebral Cortex in Progenitors and Postmitotic Neurons. Neuron 2015; 88:1192-1207. [PMID: 26671461 DOI: 10.1016/j.neuron.2015.10.045] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 08/13/2015] [Accepted: 10/13/2015] [Indexed: 11/27/2022]
Abstract
We demonstrate using conditional mutagenesis that Pbx1, with and without Pbx2(+/-) sensitization, regulates regional identity and laminar patterning of the developing mouse neocortex in cortical progenitors (Emx1-Cre) and in newly generated neurons (Nex1-Cre). Pbx1/2 mutants have three salient molecular phenotypes of cortical regional and laminar organization: hypoplasia of the frontal cortex, ventral expansion of the dorsomedial cortex, and ventral expansion of Reelin expression in the cortical plate of the frontal cortex, concomitant with an inversion of cortical layering in the rostral cortex. Molecular analyses, including PBX ChIP-seq, provide evidence that PBX promotes frontal cortex identity by repressing genes that promote dorsocaudal fate.
Collapse
Affiliation(s)
- Olga Golonzhka
- Department of Psychiatry, Neuroscience Program and the Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, San Francisco, CA 94158, USA; Acetylon Pharmaceuticals, 70 Fargo Street, Suite 205, Boston, MA 02210, USA.
| | - Alex Nord
- Departments of Neurobiology, Physiology, and Behavior and Psychiatry and Behavioral Sciences, Center for Neuroscience, University of California Davis, Davis, CA 95618, USA
| | - Paul L F Tang
- Institute for Human Genetics, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Susan Lindtner
- Department of Psychiatry, Neuroscience Program and the Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Athena R Ypsilanti
- Department of Psychiatry, Neuroscience Program and the Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Elisabetta Ferretti
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021, USA; The Danish Stem Cell Center, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Axel Visel
- Genomics Division, MS 84-171, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA 94598, USA; School of Natural Sciences, University of California, Merced, Merced, CA 95343, USA
| | - Licia Selleri
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021, USA
| | - John L R Rubenstein
- Department of Psychiatry, Neuroscience Program and the Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
91
|
En1 directs superior olivary complex neuron positioning, survival, and expression of FoxP1. Dev Biol 2015; 408:99-108. [PMID: 26542008 DOI: 10.1016/j.ydbio.2015.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/14/2015] [Accepted: 10/02/2015] [Indexed: 01/16/2023]
Abstract
Little is known about the genetic pathways and transcription factors that control development and maturation of central auditory neurons. En1, a gene expressed by a subset of developing and mature superior olivary complex (SOC) cells, encodes a homeodomain transcription factor important for neuronal development in the midbrain, cerebellum, hindbrain and spinal cord. Using genetic fate-mapping techniques, we show that all En1-lineal cells in the SOC are neurons and that these neurons are glycinergic, cholinergic and GABAergic in neurotransmitter phenotype. En1 deletion does not interfere with specification or neural fate of these cells, but does cause aberrant positioning and subsequent death of all En1-lineal SOC neurons by early postnatal ages. En1-null cells also fail to express the transcription factor FoxP1, suggesting that FoxP1 lies downstream of En1. Our data define important roles for En1 in the development and maturation of a diverse group of brainstem auditory neurons.
Collapse
|
92
|
Sullivan JM, Badimon A, Schaefer U, Ayata P, Gray J, Chung CW, von Schimmelmann M, Zhang F, Garton N, Smithers N, Lewis H, Tarakhovsky A, Prinjha RK, Schaefer A. Autism-like syndrome is induced by pharmacological suppression of BET proteins in young mice. J Exp Med 2015; 212:1771-81. [PMID: 26392221 PMCID: PMC4612093 DOI: 10.1084/jem.20151271] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/25/2015] [Indexed: 01/09/2023] Open
Abstract
Studies investigating the causes of autism spectrum disorder (ASD) point to genetic, as well as epigenetic, mechanisms of the disease. Identification of epigenetic processes that contribute to ASD development and progression is of major importance and may lead to the development of novel therapeutic strategies. Here, we identify the bromodomain and extraterminal domain-containing proteins (BETs) as epigenetic regulators of genes involved in ASD-like behaviors in mice. We found that the pharmacological suppression of BET proteins in the brain of young mice, by the novel, highly specific, brain-permeable inhibitor I-BET858 leads to selective suppression of neuronal gene expression followed by the development of an autism-like syndrome. Many of the I-BET858-affected genes have been linked to ASD in humans, thus suggesting the key role of the BET-controlled gene network in the disorder. Our studies suggest that environmental factors controlling BET proteins or their target genes may contribute to the epigenetic mechanism of ASD.
Collapse
Affiliation(s)
- Josefa M Sullivan
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ana Badimon
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Uwe Schaefer
- Laboratory of Immune Cell Epigenetics and Signaling, The Rockefeller University, New York, NY 10065
| | - Pinar Ayata
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - James Gray
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Chun-wa Chung
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Melanie von Schimmelmann
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Fan Zhang
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Neil Garton
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Nicholas Smithers
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Huw Lewis
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Alexander Tarakhovsky
- Laboratory of Immune Cell Epigenetics and Signaling, The Rockefeller University, New York, NY 10065
| | - Rab K Prinjha
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Anne Schaefer
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
93
|
Abstract
Language is a defining characteristic of the human species, but its foundations remain mysterious. Heritable disorders offer a gateway into biological underpinnings, as illustrated by the discovery that FOXP2 disruptions cause a rare form of speech and language impairment. The genetic architecture underlying language-related disorders is complex, and although some progress has been made, it has proved challenging to pinpoint additional relevant genes with confidence. Next-generation sequencing and genome-wide association studies are revolutionizing understanding of the genetic bases of other neurodevelopmental disorders, like autism and schizophrenia, and providing fundamental insights into the molecular networks crucial for typical brain development. We discuss how a similar genomic perspective, brought to the investigation of language-related phenotypes, promises to yield equally informative discoveries. Moreover, we outline how follow-up studies of genetic findings using cellular systems and animal models can help to elucidate the biological mechanisms involved in the development of brain circuits supporting language.
Collapse
Affiliation(s)
- Sarah A Graham
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands;
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands; .,Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 EN Nijmegen, The Netherlands;
| |
Collapse
|
94
|
Li X, Xiao J, Fröhlich H, Tu X, Li L, Xu Y, Cao H, Qu J, Rappold GA, Chen JG. Foxp1 regulates cortical radial migration and neuronal morphogenesis in developing cerebral cortex. PLoS One 2015; 10:e0127671. [PMID: 26010426 PMCID: PMC4444005 DOI: 10.1371/journal.pone.0127671] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 04/17/2015] [Indexed: 11/30/2022] Open
Abstract
FOXP1 is a member of FOXP subfamily transcription factors. Mutations in FOXP1 gene have been found in various development-related cognitive disorders. However, little is known about the etiology of these symptoms, and specifically the function of FOXP1 in neuronal development. Here, we report that suppression of Foxp1 expression in mouse cerebral cortex led to a neuronal migration defect, which was rescued by overexpression of Foxp1. Mice with Foxp1 knockdown exhibited ectopic neurons in deep layers of the cortex postnatally. The neuronal differentiation of Foxp1-downregulated cells was normal. However, morphological analysis showed that the neurons with Foxp1 deficiency had an inhibited axonal growth in vitro and a weakened transition from multipolar to bipolar in vivo. Moreover, we found that the expression of Foxp1 modulated the dendritic maturation of neurons at a late postnatal date. Our results demonstrate critical roles of Foxp1 in the radial migration and morphogenesis of cortical neurons during development. This study may shed light on the complex relationship between neuronal development and the related cognitive disorders.
Collapse
Affiliation(s)
- Xue Li
- Key Laboratory of Visual Science, National Ministry of Health, and School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Jian Xiao
- Key Laboratory of Visual Science, National Ministry of Health, and School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Henning Fröhlich
- Department of Human Molecular Genetics, University of Heidelberg, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Xiaomeng Tu
- Key Laboratory of Visual Science, National Ministry of Health, and School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Lianlian Li
- Key Laboratory of Visual Science, National Ministry of Health, and School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Yue Xu
- Key Laboratory of Visual Science, National Ministry of Health, and School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Huateng Cao
- Key Laboratory of Visual Science, National Ministry of Health, and School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Jia Qu
- Key Laboratory of Visual Science, National Ministry of Health, and School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, University of Heidelberg, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Jie-Guang Chen
- Key Laboratory of Visual Science, National Ministry of Health, and School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| |
Collapse
|
95
|
Lozano R, Vino A, Lozano C, Fisher SE, Deriziotis P. A de novo FOXP1 variant in a patient with autism, intellectual disability and severe speech and language impairment. Eur J Hum Genet 2015; 23:1702-7. [PMID: 25853299 DOI: 10.1038/ejhg.2015.66] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 02/05/2015] [Accepted: 02/26/2015] [Indexed: 12/19/2022] Open
Abstract
FOXP1 (forkhead box protein P1) is a transcription factor involved in the development of several tissues, including the brain. An emerging phenotype of patients with protein-disrupting FOXP1 variants includes global developmental delay, intellectual disability and mild to severe speech/language deficits. We report on a female child with a history of severe hypotonia, autism spectrum disorder and mild intellectual disability with severe speech/language impairment. Clinical exome sequencing identified a heterozygous de novo FOXP1 variant c.1267_1268delGT (p.V423Hfs*37). Functional analyses using cellular models show that the variant disrupts multiple aspects of FOXP1 activity, including subcellular localization and transcriptional repression properties. Our findings highlight the importance of performing functional characterization to help uncover the biological significance of variants identified by genomics approaches, thereby providing insight into pathways underlying complex neurodevelopmental disorders. Moreover, our data support the hypothesis that de novo variants represent significant causal factors in severe sporadic disorders and extend the phenotype seen in individuals with FOXP1 haploinsufficiency.
Collapse
Affiliation(s)
- Reymundo Lozano
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Department of Pediatrics, UC Davis Medical Center, Sacramento, CA, USA
| | - Arianna Vino
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Cristina Lozano
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Department of Pediatrics, UC Davis Medical Center, Sacramento, CA, USA
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Pelagia Deriziotis
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| |
Collapse
|
96
|
Walker MP, Stopford CM, Cederlund M, Fang F, Jahn C, Rabinowitz AD, Goldfarb D, Graham DM, Yan F, Deal AM, Fedoriw Y, Richards KL, Davis IJ, Weidinger G, Damania B, Major MB. FOXP1 potentiates Wnt/β-catenin signaling in diffuse large B cell lymphoma. Sci Signal 2015; 8:ra12. [PMID: 25650440 DOI: 10.1126/scisignal.2005654] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transcription factor FOXP1 (forkhead box protein P1) is a master regulator of stem and progenitor cell biology. In diffuse large B cell lymphoma (DLBCL), copy number amplifications and chromosomal translocations result in overexpression of FOXP1. Increased abundance of FOXP1 in DLBCL is a predictor of poor prognosis and resistance to therapy. We developed a genome-wide, mass spectrometry-coupled, gain-of-function genetic screen, which revealed that FOXP1 potentiates β-catenin-dependent, Wnt-dependent gene expression. Gain- and loss-of-function studies in cell models and zebrafish confirmed that FOXP1 was a general and conserved enhancer of Wnt signaling. In a Wnt-dependent fashion, FOXP1 formed a complex with β-catenin, TCF7L2 (transcription factor 7-like 2), and the acetyltransferase CBP [CREB (adenosine 3',5'-monophosphate response element-binding protein)-binding protein], and this complex bound the promoters of Wnt target genes. FOXP1 promoted the acetylation of β-catenin by CBP, and acetylation was required for FOXP1-mediated potentiation of β-catenin-dependent transcription. In DLBCL, we found that FOXP1 promoted sensitivity to Wnt pathway inhibitors, and knockdown of FOXP1 or blocking β-catenin transcriptional activity slowed xenograft tumor growth. These data connect excessive FOXP1 with β-catenin-dependent signal transduction and provide a molecular rationale for Wnt-directed therapy in DLBCL.
Collapse
Affiliation(s)
- Matthew P Walker
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Charles M Stopford
- Division of Microbiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27516-7361, USA
| | - Maria Cederlund
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Fang Fang
- Carolina Center for Genome Sciences, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Christopher Jahn
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Alex D Rabinowitz
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Dennis Goldfarb
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3175, USA
| | - David M Graham
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Feng Yan
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Allison M Deal
- UNC Lineberger Comprehensive Cancer Center Biostatistics Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Yuri Fedoriw
- Department of Pathology and Laboratory, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Kristy L Richards
- Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27516-7361, USA
| | - Ian J Davis
- Carolina Center for Genome Sciences, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Gilbert Weidinger
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Blossom Damania
- Division of Microbiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27516-7361, USA
| | - Michael B Major
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA. Division of Microbiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27516-7361, USA.
| |
Collapse
|
97
|
Walker MP, Major MB, VanHook AM. Science Signaling
Podcast: 3 February 2015. Sci Signal 2015. [DOI: 10.1126/scisignal.aaa7112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Wnt signaling inhibitors could be used to treat a common type of lymphoma.
Collapse
Affiliation(s)
- Matthew P. Walker
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Michael B. Major
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
- Division of Microbiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27516-7361, USA
| | - Annalisa M. VanHook
- Web Editor, Science Signaling, American Association for the Advancement of Science, 1200 New York Avenue, NW, Washington, DC 20005, USA
| |
Collapse
|