MEIS2 Gene Is Responsible for Intellectual Disability, Cardiac

Total Page:16

File Type:pdf, Size:1020Kb

MEIS2 Gene Is Responsible for Intellectual Disability, Cardiac European Journal of Medical Genetics 63 (2020) 103627 Contents lists available at ScienceDirect European Journal of Medical Genetics journal homepage: www.elsevier.com/locate/ejmg MEIS2 gene is responsible for intellectual disability, cardiac defects and a T distinct facial phenotype Annarita Gilibertia,b,1, Aurora Curròa,b,1, Filomena Tiziana Papaa, Elisa Frullantia, ∗ Francesca Ariania,b, Gianni Coriolanic, Salvatore Grossoc, Alessandra Renieria,b, , Francesca Maria,b a Medical Genetics, University of Siena, Siena, Italy b Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy c Clinical Pediatrics, Department of Molecular Medicine and Development, University of Siena, Siena, Italy ARTICLE INFO ABSTRACT Keywords: Myeloid ecotropic insertion site 2 (MEIS2) gene, encoding a homeodomain-containing transcription factor, has Ventricular septal defect been recently related to syndromic intellectual disability with cleft palate and cardiac defects. Here, we present a Intellectual disability male patient, aged 10, with cardiac defects, intellectual disability, facial dysmorphisms and gastroesophageal Exome analysis reflux. Whole exome sequencing revealed a novel de novo nonsense mutation inthe MEIS2 gene. This patient MEIS2 represents another reported case with a de novo MEIS2 point mutation and helps to characterize a distinct facial phenotype consisting in low anterior hairline, thin eyebrows, anteverted nares, hypoplastic alae nasi, and M- shape upper lip. Furthermore, these data confirm the role of this gene in cardiac, nervous system development and gastrointestinal function. 1. Introduction novo MEIS2 point mutation and propose that a distinct facial phenotype is present in these patients making the MEIS2-related condition a dis- Myeloid ecotropic insertion site 2 (MEIS2) gene (OMIM # 600987) tinct and recognizable neurodevelopmental disorder. encodes a homeodomain-containing transcription factor belonging to the TALE (three amino acid loop extension) highly conserved super- 2. Materials and methods family of transcription regulators. Recent studies identified MEIS2 as a gene essential for cranial and cardiac neural crest development 2.1. Sample and DNA extraction (Machon et al., 2015). The first evidences of MEIS2 involvement in syndromic ID date back to 2007 when a unique case with intellectual Parents provided and signed a written informed consent at the disability (ID), atrial septal defect type secundum and cleft palate and a Medical Genetics Unit of the Azienda Ospedaliera Universitaria Senese, 15q14 microdeletion including MEIS2 was reported (Erdogan et al., Siena, Italy, for exome sequencing analysis, clinical data usage, and the 2007). Subsequently additional few patients with a copy number var- use of DNA samples from the tested individuals for both diagnostic and iation including MEIS2, either partially or totally together with flanking research purposes. Genomic DNA was extracted from EDTA peripheral genes, were reported, indicating that MEIS2 was the phenotype causa- blood samples using MagCore HF16 (Diatech Lab Line, Jesi, Ancona, tive gene (Chen et al., 2008; Crowley et al., 2010; Johansson et al., Italy). 2014; Gambin et al., 2017; Shimojima et al., 2017; Brunetti-Pierri et al., 2008; Conte et al., 2016). Very recently, additional de novo MEIS2 2.2. Whole Exome Sequencing point mutations have been described in patients with ID, palate ab- normalities, congenital heart defect, and facial dysmorphisms (Louw Exome sequencing was performed on genomic DNA samples of et al., 2015; Fujita et al., 2016; Srivastava et al., 2018; Douglas et al., proband and both parents. The library preparation was performed using 2018; Verheije et al., 2018). Here, we present another patient with a de the Ion AmpliSeq™ Exome Kit that allows to target ∼33 Mb of coding ∗ Corresponding author. Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy. E-mail address: [email protected] (A. Renieri). 1 These authors contributed equally to the work. https://doi.org/10.1016/j.ejmg.2019.01.017 Received 7 August 2018; Received in revised form 25 January 2019; Accepted 29 January 2019 Available online 05 February 2019 1769-7212/ © 2019 Elsevier Masson SAS. All rights reserved. A. Giliberti, et al. European Journal of Medical Genetics 63 (2020) 103627 Fig. 1. Pictures of our patient and the first two reported patients with a MEIS2 point mutation. A) Present patient B) Patient reported by Fujita et al. C) Patient reported by Louw et al. Permission for reproduction of the pictures of the two already published cases has been asked to the Publishing Company. exons plus 15 Mb of flanking regions for a total of ∼58 Mb, intotal specific primer pairs (Forward 5′-AGACATCACACTGTTGCTCTT-3’; more than 97% of the coding regions as described by Consensus Coding Reverse 5-’AGACCCCAAAAGAGACGGAG-3′) and PCR products were Sequences (CCDS) annotation, using 12 primer pools for highly specific sequenced employing ABIPRISM3130 Genetic Analyzer (Applied enrichment of exons within the human genome. Taking advantage of a Biosystems, Foster City, CA, USA) and data were analyzed with barcode system, three samples were loaded together in a single run and Sequencher software V.4.9 (Gene Codes, Ann Arbor, USA). sequenced. Exome sequencing was performed using the Life Technologies Ion Proton sequencer (Life Technologies). Data analysis 2.4. Mutation nomenclature was performed with Torrent SuitetTM Software v3.6.2. The provider generated at least 30X effective mean depth per sample. Using specific The mutation is described according to Human Genome Variations parameters we obtained removing adaptors contamination and low Society (HGVS) (den Dunnen, 2016 #195). Nucleotide numbers are quality sequences, so the total amount of clean data was mapped to the derived from the cDNA sequence of MEIS2 (GenBank accession no. NM_ UCSC/hg19 reference genome. Indel and variants calls were made using 170674.4). GATK version 2.7 (and recommended parameters) (McKenna et al., 2010)and then the variants were also annotated against external da- tasets including 1000 genomes and dbSNP. 3. Results Prioritization of the variants was obtained selecting frameshift, splice, stopgain or stoploss mutations, missense variants predicted to be 3.1. Clinical description damaging by CADD-phred prediction tools and variants with minor allele frequency (MAF) < 0,01. Here we present a caucasian male patient, aged 10, evaluated in our Unit because of intellectual disability. Parents are not consanguineous and aged 28 years (mother) and 29 years (father) at the time of de- 2.3. Sanger sequencing livery. He was born at 37 gws with cesarean section; his weight was 2540 g (−0.9 SD), his length was 47 cm (−0.53 SD), his occipital Sanger sequencing was performed in the proband and his parents. frontal circumference (OFC) was 31,8 cm (−1.58 SD), and the APGAR DNA was amplified by PCR on exon 6 of MEIS2 (NM_170674.4) using index was 8–10. He had a history of ventricular and atrial septal defect 2 A. Giliberti, et al. European Journal of Medical Genetics 63 (2020) 103627 Fig. 2. MEIS2 point mutations. Upper panel (A): Mutation confirmation and segregation analysis by Sanger sequencing. Lower panel (B): Scheme oftheMEIS2 protein with the localization of the mutation reported in this paper. that solved without surgery and undescended right testicle. He had 4. Discussion psychomotor delay: he reached trunk control at 12 months, he was able to walk after 18 months, to speak at 5 years; he got sphincter control We report on a patient carrying a de novo nonsense mutation in after the age of 5 years. We evaluated the patient for the first time when MEIS2, a gene previously associated with syndromic ID (Johansson he was 8 years old: he was 133 cm tall (0.06 SD), his weight was et al., 2014; Louw et al., 2015; Douglas et al., 2018; Verheije et al., 34,4 Kg (1.03 SD) and his OFC was 55 cm (2 SD); he had large forehead, 2018). Cardiac defects, palate anomalies and gastroesophageal dis- low frontal hairline, thick hair, thin and laterally extended eyebrows, turbances are among the most frequent comorbidities (Johansson et al., large nasal tip with anteverted and hypoplastic nostrils, M-shaped 2014; Louw et al., 2015; Fujita et al., 2016; Douglas et al., 2018; upper lip, high palate, tapering fingers, sandal gap bilaterally (Fig. 1A). Verheije et al., 2018). Till now 47 unrelated patients with MEIS2 mu- His language was very poor and repetitive, and he had stereotyping tations have been described. The majority of these patients have been movements. He had a IQ of 45 (according to a WISC-IV scale), and he identified by array-CGH analysis and carried different microdeletions was diagnosed with Autism Spectrum Disorder (ASD). He also pre- involving MEIS2. In four patients the microdeletion either interrupted sented stereotypic hand and trunk movements. In particular, he showed the gene or involved uniquely the MEIS2 gene, pointing to this gene as clenched fists held close while rocking the trunk. His EEG had nopat- the major gene responsible for the phenotype. tern anomalies; the most recent brain MRI detected ectasia and gliosis Here, we describe another patient bearing a MEIS2 point mutation of Virchow-Robin areas. He had no anomalies at US evaluation of the (Table 1). abdomen nor at an evaluation of the anterior eye chamber and fundus Pathogenic sequence changes reported to date include six truncating oculi. A cardiac US confirmed the presence of VSD. He was previously mutation, six missense mutation, three splice variants and one in-frame investigated for Fragile-X, with a negative result. A previous array-CGH deletion (Fujita et al., 2016; Srivastava et al., 2018; Louw et al., 2015; analysis revealed a small 19q13.12 microdeletion paternally inherited Douglas et al., 2018; Verheije et al., 2018). All patients except one of not including known disease genes, specifically arr [GRCh37] 19q13.12 those reported by Verheije et al., who was diagnosed by targeted se- (42595081_42744350)×1. quencing of MEIS2, have been identified by whole exome analysis, suggesting that the phenotype associated with MEIS2 point mutations is not yet easily recognizable (Verheije et al., 2018).
Recommended publications
  • De Novo MEIS2 Mutation Causes Syndromic Developmental Delay with Persistent Gastro-Esophageal Reflux
    Journal of Human Genetics (2016) 61, 835–838 & 2016 The Japan Society of Human Genetics All rights reserved 1434-5161/16 www.nature.com/jhg SHORT COMMUNICATION De novo MEIS2 mutation causes syndromic developmental delay with persistent gastro-esophageal reflux Atsushi Fujita1, Bertrand Isidor2,3, Hugues Piloquet4, Pierre Corre5, Nobuhiko Okamoto6, Mitsuko Nakashima1, Yoshinori Tsurusaki1, Hirotomo Saitsu1, Noriko Miyake1 and Naomichi Matsumoto1 MEIS2 aberrations are considered to be the cause of intellectual disability, cleft palate and cardiac septal defect, as MEIS2 copy number variation is often observed with these phenotypes. To our knowledge, only one nucleotide-level change— specifically, an in-frame MEIS2 deletion—has so far been reported. Here, we report a female patient with a de novo nonsense mutation (c.611C4G, p.Ser204*) in MEIS2. She showed severe intellectual disability, moderate motor/verbal developmental delay, cleft palate, cardiac septal defect, hypermetropia, severe feeding difficulties with gastro-esophageal reflux and constipation. By reviewing this patient and previous patients with MEIS2 point mutations, we found that feeding difficulty with gastro-esophageal reflux appears to be one of the core clinical features of MEIS2 haploinsufficiency, in addition to intellectual disability, cleft palate and cardiac septal defect. Journal of Human Genetics (2016) 61, 835–838; doi:10.1038/jhg.2016.54; published online 26 May 2016 INTRODUCTION at the homeodomain may interfere with DNA binding. Moreover, Meis homeobox 2 (MEIS2;alsoknownasMRG1, NM_170677.4) this female patient also presented with various anomalous features encodes a homeobox (HOX) protein belonging to the three amino and developmental abnormalities. Here, we report on another acid loop extension (TALE) superfamily.
    [Show full text]
  • Detailed Review Paper on Retinoid Pathway Signalling
    1 1 Detailed Review Paper on Retinoid Pathway Signalling 2 December 2020 3 2 4 Foreword 5 1. Project 4.97 to develop a Detailed Review Paper (DRP) on the Retinoid System 6 was added to the Test Guidelines Programme work plan in 2015. The project was 7 originally proposed by Sweden and the European Commission later joined the project as 8 a co-lead. In 2019, the OECD Secretariat was added to coordinate input from expert 9 consultants. The initial objectives of the project were to: 10 draft a review of the biology of retinoid signalling pathway, 11 describe retinoid-mediated effects on various organ systems, 12 identify relevant retinoid in vitro and ex vivo assays that measure mechanistic 13 effects of chemicals for development, and 14 Identify in vivo endpoints that could be added to existing test guidelines to 15 identify chemical effects on retinoid pathway signalling. 16 2. This DRP is intended to expand the recommendations for the retinoid pathway 17 included in the OECD Detailed Review Paper on the State of the Science on Novel In 18 vitro and In vivo Screening and Testing Methods and Endpoints for Evaluating 19 Endocrine Disruptors (DRP No 178). The retinoid signalling pathway was one of seven 20 endocrine pathways considered to be susceptible to environmental endocrine disruption 21 and for which relevant endpoints could be measured in new or existing OECD Test 22 Guidelines for evaluating endocrine disruption. Due to the complexity of retinoid 23 signalling across multiple organ systems, this effort was foreseen as a multi-step process.
    [Show full text]
  • Prox1regulates the Subtype-Specific Development of Caudal Ganglionic
    The Journal of Neuroscience, September 16, 2015 • 35(37):12869–12889 • 12869 Development/Plasticity/Repair Prox1 Regulates the Subtype-Specific Development of Caudal Ganglionic Eminence-Derived GABAergic Cortical Interneurons X Goichi Miyoshi,1 Allison Young,1 Timothy Petros,1 Theofanis Karayannis,1 Melissa McKenzie Chang,1 Alfonso Lavado,2 Tomohiko Iwano,3 Miho Nakajima,4 Hiroki Taniguchi,5 Z. Josh Huang,5 XNathaniel Heintz,4 Guillermo Oliver,2 Fumio Matsuzaki,3 Robert P. Machold,1 and Gord Fishell1 1Department of Neuroscience and Physiology, NYU Neuroscience Institute, Smilow Research Center, New York University School of Medicine, New York, New York 10016, 2Department of Genetics & Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, 3Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan, 4Laboratory of Molecular Biology, Howard Hughes Medical Institute, GENSAT Project, The Rockefeller University, New York, New York 10065, and 5Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724 Neurogliaform (RELNϩ) and bipolar (VIPϩ) GABAergic interneurons of the mammalian cerebral cortex provide critical inhibition locally within the superficial layers. While these subtypes are known to originate from the embryonic caudal ganglionic eminence (CGE), the specific genetic programs that direct their positioning, maturation, and integration into the cortical network have not been eluci- dated. Here, we report that in mice expression of the transcription factor Prox1 is selectively maintained in postmitotic CGE-derived cortical interneuron precursors and that loss of Prox1 impairs the integration of these cells into superficial layers. Moreover, Prox1 differentially regulates the postnatal maturation of each specific subtype originating from the CGE (RELN, Calb2/VIP, and VIP).
    [Show full text]
  • A Conserved Tissue-Specific Homeodomain-Less Isoform of MEIS1 Is Downregulated in Colorectal Cancer
    Thomas Jefferson University Jefferson Digital Commons Department of Microbiology and Immunology Faculty Papers Department of Microbiology and Immunology 8-17-2011 A conserved tissue-specific homeodomain-less isoform of MEIS1 is downregulated in colorectal cancer. Richard C Crist Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia Jacquelyn J Roth Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia Scott A Waldman Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia Arthur M Buchberg Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia Follow this and additional works at: https://jdc.jefferson.edu/mifp Part of the Gastroenterology Commons, Medical Genetics Commons, Medical Microbiology Commons, Oncology Commons, and the Pathology Commons Let us know how access to this document benefits ouy Recommended Citation Crist, Richard C; Roth, Jacquelyn J; Waldman, Scott A; and Buchberg, Arthur M, "A conserved tissue-specific homeodomain-less isoform of MEIS1 is downregulated in colorectal cancer." (2011). Department of Microbiology and Immunology Faculty Papers. Paper 25. https://jdc.jefferson.edu/mifp/25 This Article is brought to you for free and open access by the Jefferson Digital Commons. The Jefferson Digital Commons is a service of Thomas Jefferson University's Center for Teaching and Learning (CTL). The Commons is a showcase for Jefferson books and journals, peer-reviewed scholarly publications, unique historical collections from the University archives, and teaching tools. The Jefferson Digital Commons allows researchers and interested readers anywhere in the world to learn about and keep up to date with Jefferson scholarship. This article has been accepted for inclusion in Department of Microbiology and Immunology Faculty Papers by an authorized administrator of the Jefferson Digital Commons.
    [Show full text]
  • The Role of Meis1 in Hematopoietic Development Mi Cai Washington University in St
    Washington University in St. Louis Washington University Open Scholarship All Theses and Dissertations (ETDs) 8-31-2012 The Role of Meis1 in Hematopoietic Development Mi Cai Washington University in St. Louis Follow this and additional works at: https://openscholarship.wustl.edu/etd Recommended Citation Cai, Mi, "The Role of Meis1 in Hematopoietic Development" (2012). All Theses and Dissertations (ETDs). 944. https://openscholarship.wustl.edu/etd/944 This Dissertation is brought to you for free and open access by Washington University Open Scholarship. It has been accepted for inclusion in All Theses and Dissertations (ETDs) by an authorized administrator of Washington University Open Scholarship. For more information, please contact [email protected]. WASHINGTON UNIVERSITY IN ST. LOUIS Division of Biology and Biomedical Sciences Molecular Cell Biology Dissertation Examination Committee: Kenneth M. Murphy, Chair David C. Beebe Deepta Bhattacharya Raphael Kopan Kristen Kroll David M. Ornitz The Role of Meis1 in Hematopoietic Development By Mi Cai A dissertation presented to the Graduate School of Arts and Sciences of Washington University in partial fulfillment of the requirements for the degree of Doctor of Philosophy August 2012 Saint Louis, Missouri ABSTRACT OF THE DISSERTATION The Role of Meis1 in Hematopoietic Development By Mi Cai Doctor of Philosophy in Biology and Biomedical Sciences Molecular Cell Biology Washington University in St. Louis, 2012 Professor Kenneth M. Murphy, Chair Previous work has identified Mesp1 as an important regulator of the epithelial- mesenchymal transition (EMT) and of cardiovascular cell fate in differentiating embryonic stem cells (ESCs). To understand the molecular mechanisms underlying the actions of Mesp1, we sought to identify transcription targets of Mesp1.
    [Show full text]
  • A Single-Cell Transcriptome Atlas of the Mouse Glomerulus
    RAPID COMMUNICATION www.jasn.org A Single-Cell Transcriptome Atlas of the Mouse Glomerulus Nikos Karaiskos,1 Mahdieh Rahmatollahi,2 Anastasiya Boltengagen,1 Haiyue Liu,1 Martin Hoehne ,2 Markus Rinschen,2,3 Bernhard Schermer,2,4,5 Thomas Benzing,2,4,5 Nikolaus Rajewsky,1 Christine Kocks ,1 Martin Kann,2 and Roman-Ulrich Müller 2,4,5 Due to the number of contributing authors, the affiliations are listed at the end of this article. ABSTRACT Background Three different cell types constitute the glomerular filter: mesangial depending on cell location relative to the cells, endothelial cells, and podocytes. However, to what extent cellular heteroge- glomerular vascular pole.3 Because BP ad- neity exists within healthy glomerular cell populations remains unknown. aptation and mechanoadaptation of glo- merular cells are key determinants of kidney Methods We used nanodroplet-based highly parallel transcriptional profiling to function and dysregulated in kidney disease, characterize the cellular content of purified wild-type mouse glomeruli. we tested whether glomerular cell type sub- Results Unsupervised clustering of nearly 13,000 single-cell transcriptomes identi- sets can be identified by single-cell RNA fied the three known glomerular cell types. We provide a comprehensive online sequencing in wild-type glomeruli. This atlas of gene expression in glomerular cells that can be queried and visualized using technique allows for high-throughput tran- an interactive and freely available database. Novel marker genes for all glomerular scriptome profiling of individual cells and is cell types were identified and supported by immunohistochemistry images particularly suitable for identifying novel obtained from the Human Protein Atlas.
    [Show full text]
  • Full-Text.Pdf
    Systematic Evaluation of Genes and Genetic Variants Associated with Type 1 Diabetes Susceptibility This information is current as Ramesh Ram, Munish Mehta, Quang T. Nguyen, Irma of September 23, 2021. Larma, Bernhard O. Boehm, Flemming Pociot, Patrick Concannon and Grant Morahan J Immunol 2016; 196:3043-3053; Prepublished online 24 February 2016; doi: 10.4049/jimmunol.1502056 Downloaded from http://www.jimmunol.org/content/196/7/3043 Supplementary http://www.jimmunol.org/content/suppl/2016/02/19/jimmunol.150205 Material 6.DCSupplemental http://www.jimmunol.org/ References This article cites 44 articles, 5 of which you can access for free at: http://www.jimmunol.org/content/196/7/3043.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 23, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Systematic Evaluation of Genes and Genetic Variants Associated with Type 1 Diabetes Susceptibility Ramesh Ram,*,† Munish Mehta,*,† Quang T.
    [Show full text]
  • Mammalian PRC1 Complexes: Compositional Complexity and Diverse Molecular Mechanisms
    International Journal of Molecular Sciences Review Mammalian PRC1 Complexes: Compositional Complexity and Diverse Molecular Mechanisms Zhuangzhuang Geng 1 and Zhonghua Gao 1,2,3,* 1 Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA; [email protected] 2 Penn State Hershey Cancer Institute, Hershey, PA 17033, USA 3 The Stem Cell and Regenerative Biology Program, Penn State College of Medicine, Hershey, PA 17033, USA * Correspondence: [email protected] Received: 6 October 2020; Accepted: 5 November 2020; Published: 14 November 2020 Abstract: Polycomb group (PcG) proteins function as vital epigenetic regulators in various biological processes, including pluripotency, development, and carcinogenesis. PcG proteins form multicomponent complexes, and two major types of protein complexes have been identified in mammals to date, Polycomb Repressive Complexes 1 and 2 (PRC1 and PRC2). The PRC1 complexes are composed in a hierarchical manner in which the catalytic core, RING1A/B, exclusively interacts with one of six Polycomb group RING finger (PCGF) proteins. This association with specific PCGF proteins allows for PRC1 to be subdivided into six distinct groups, each with their own unique modes of action arising from the distinct set of associated proteins. Historically, PRC1 was considered to be a transcription repressor that deposited monoubiquitylation of histone H2A at lysine 119 (H2AK119ub1) and compacted local chromatin. More recently, there is increasing evidence that demonstrates the transcription activation role of PRC1. Moreover, studies on the higher-order chromatin structure have revealed a new function for PRC1 in mediating long-range interactions. This provides a different perspective regarding both the transcription activation and repression characteristics of PRC1.
    [Show full text]
  • BMC Biology Biomed Central
    BMC Biology BioMed Central Research article Open Access Classification and nomenclature of all human homeobox genes PeterWHHolland*†1, H Anne F Booth†1 and Elspeth A Bruford2 Address: 1Department of Zoology, University of Oxford, South Parks Road, Oxford, OX1 3PS, UK and 2HUGO Gene Nomenclature Committee, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK Email: Peter WH Holland* - [email protected]; H Anne F Booth - [email protected]; Elspeth A Bruford - [email protected] * Corresponding author †Equal contributors Published: 26 October 2007 Received: 30 March 2007 Accepted: 26 October 2007 BMC Biology 2007, 5:47 doi:10.1186/1741-7007-5-47 This article is available from: http://www.biomedcentral.com/1741-7007/5/47 © 2007 Holland et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Background: The homeobox genes are a large and diverse group of genes, many of which play important roles in the embryonic development of animals. Increasingly, homeobox genes are being compared between genomes in an attempt to understand the evolution of animal development. Despite their importance, the full diversity of human homeobox genes has not previously been described. Results: We have identified all homeobox genes and pseudogenes in the euchromatic regions of the human genome, finding many unannotated, incorrectly annotated, unnamed, misnamed or misclassified genes and pseudogenes.
    [Show full text]
  • A Grainyhead-Like 2/Ovo-Like 2 Pathway Regulates Renal Epithelial Barrier Function and Lumen Expansion
    BASIC RESEARCH www.jasn.org A Grainyhead-Like 2/Ovo-Like 2 Pathway Regulates Renal Epithelial Barrier Function and Lumen Expansion † ‡ | Annekatrin Aue,* Christian Hinze,* Katharina Walentin,* Janett Ruffert,* Yesim Yurtdas,*§ | Max Werth,* Wei Chen,* Anja Rabien,§ Ergin Kilic,¶ Jörg-Dieter Schulzke,** †‡ Michael Schumann,** and Kai M. Schmidt-Ott* *Max Delbrueck Center for Molecular Medicine, Berlin, Germany; †Experimental and Clinical Research Center, and Departments of ‡Nephrology, §Urology, ¶Pathology, and **Gastroenterology, Charité Medical University, Berlin, Germany; and |Berlin Institute of Urologic Research, Berlin, Germany ABSTRACT Grainyhead transcription factors control epithelial barriers, tissue morphogenesis, and differentiation, but their role in the kidney is poorly understood. Here, we report that nephric duct, ureteric bud, and collecting duct epithelia express high levels of grainyhead-like homolog 2 (Grhl2) and that nephric duct lumen expansion is defective in Grhl2-deficient mice. In collecting duct epithelial cells, Grhl2 inactivation impaired epithelial barrier formation and inhibited lumen expansion. Molecular analyses showed that GRHL2 acts as a transcrip- tional activator and strongly associates with histone H3 lysine 4 trimethylation. Integrating genome-wide GRHL2 binding as well as H3 lysine 4 trimethylation chromatin immunoprecipitation sequencing and gene expression data allowed us to derive a high-confidence GRHL2 target set. GRHL2 transactivated a group of genes including Ovol2, encoding the ovo-like 2 zinc finger transcription factor, as well as E-cadherin, claudin 4 (Cldn4), and the small GTPase Rab25. Ovol2 induction alone was sufficient to bypass the requirement of Grhl2 for E-cadherin, Cldn4,andRab25 expression. Re-expression of either Ovol2 or a combination of Cldn4 and Rab25 was sufficient to rescue lumen expansion and barrier formation in Grhl2-deficient collecting duct cells.
    [Show full text]
  • The BAF45D Protein Is Preferentially Expressed in Adult Neurogenic Zones and in Neurons and May Be Required for Retinoid Acid Induced PAX6 Expression
    ORIGINAL RESEARCH published: 06 November 2017 doi: 10.3389/fnana.2017.00094 The BAF45D Protein Is Preferentially Expressed in Adult Neurogenic Zones and in Neurons and May Be Required for Retinoid Acid Induced PAX6 Expression Chao Liu 1, 2, 3*†, Ruyu Sun 1, 2, 3†, Jian Huang 4†, Dijuan Zhang 1, 2, 3, Dake Huang 1, Weiqin Qi 1, Shenghua Wang 1, 2, 3, Fenfen Xie 1, 2, 3, Yuxian Shen 1 and Cailiang Shen 4* 1 School of Basic Medical Sciences, Anhui Medical University, Hefei, China, 2 Department of Histology and Embryology, Anhui Medical University, Hefei, China, 3 Institute of Stem Cell and Tissue Engineering, Anhui Medical University, Hefei, China, 4 Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China Adult neurogenesis is important for the development of regenerative therapies for human diseases of the central nervous system (CNS) through the recruitment of adult neural stem cells (NSCs). NSCs are characterized by the capacity to generate neurons, Edited by: Daniel A. Peterson, astrocytes, and oligodendrocytes. To identify key factors involved in manipulating the Rosalind Franklin University of adult NSC neurogenic fate thus has crucial implications for the clinical application. Here, Medicine and Science, United States we report that BAF45D is expressed in the subgranular zone (SGZ) of the dentate gyrus, Reviewed by: Richard S. Nowakowski, the subventricular zone (SVZ) of the lateral ventricle, and the central canal (CC) of the Florida State University College of adult spinal cord. Coexpression of BAF45D with glial fibrillary acidic protein (GFAP), a Medicine, United States radial glial like cell marker protein, was identified in the SGZ, the SVZ and the adult spinal Tobias David Merson, Australian Regenerative Medicine cord CC.
    [Show full text]
  • Stage-Dependent Differential Gene Expression Profiles of Cranial
    bioRxiv preprint doi: https://doi.org/10.1101/432302; this version posted October 1, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 2 3 4 Stage-Dependent Differential Gene Expression Profiles of Cranial 5 Neural Crest Cells Derived from Mouse Induced Pluripotent Stem 6 Cells 7 8 9 Ayano Odashima1¶, Shoko Onodera2, Akiko Saito2, Takashi Nakamura2, Yuuki Ogihara3, 10 Tatsuya Ichinohe3, Toshifumi Azuma1,2* 11 12 13 1 Department of Oral Health Science Center, Tokyo Dental College, Tokyo, Japan 14 15 2 Department of Biochemistry, Tokyo Dental College, Tokyo, Japan 16 17 3 Department of Dental Anesthesiology, Tokyo Dental College, Tokyo, Japan 18 19 ¶These authors contributed equally to this work. 20 *Corresponding author 21 E-mail [email protected] 22 1 bioRxiv preprint doi: https://doi.org/10.1101/432302; this version posted October 1, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 23 Abstract 24 Cranial neural crest cells (cNCCs) comprise a multipotent population of cells that migrate into 25 the pharyngeal arches of the vertebrate embryo and differentiate into a broad range of derivatives 26 of the craniofacial organs. Consequently, migrating cNCCs are considered as one of the most 27 attractive candidate sources of cells for regenerative medicine.
    [Show full text]