Influence of Sex and Genetic Variability on Expression of X-Linked Genes In

Total Page:16

File Type:pdf, Size:1020Kb

Influence of Sex and Genetic Variability on Expression of X-Linked Genes In CORE Metadata, citation and similar papers at core.ac.uk Provided by Elsevier - Publisher Connector Genomics 98 (2011) 320–326 Contents lists available at ScienceDirect Genomics journal homepage: www.elsevier.com/locate/ygeno Influence of sex and genetic variability on expression of X-linked genes in human monocytes Raphaële Castagné a, Tanja Zeller b,1, Maxime Rotival a, Silke Szymczak c, Vinh Truong a, Arne Schillert c, David-Alexandre Trégouët a, Thomas Münzel b, Andreas Ziegler c, François Cambien a, Stefan Blankenberg b,1, Laurence Tiret a,⁎ a INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, 91 Bd de l'Hôpital 75013, Paris, France b II. Medizinische Klinik und Poliklinik, Johannes-Gutenberg Universität Mainz, Universitätsmedizin, Langenbeckstraße 1, 55131 Mainz, Germany c Institut für Medizinische Biometrie und Statistik, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany article info abstract Article history: In humans, the fraction of X-linked genes with higher expression in females has been estimated to be 5% from Received 12 April 2011 microarray studies, a proportion lower than the 25% of genes thought to escape X inactivation. We analyzed Accepted 28 June 2011 715 X-linked transcripts in circulating monocytes from 1,467 subjects and found an excess of female-biased Available online 6 July 2011 − transcripts on the X compared to autosomes (9.4% vs 5.5%, pb2×10 5). Among the genes not previously known to escape inactivation, the most significant one was EFHC2 whose 20% of variability was explained by Keywords: sex. We also investigated cis expression quantitative trait loci (eQTLs) by analyzing 15,703 X-linked SNPs. The X chromosome inactivation Gene expression frequency and magnitude of X-linked cis eQTLs were quite similar in males and females. Few genes exhibited a Expression Quantitative Trait Locus stronger genetic effect in females than in males (ARSD, DCX, POLA1 and ITM2A). These genes would deserve Gender further investigation since they may contribute to sex pathophysiological differences. © 2011 Elsevier Inc. All rights reserved. 1. Introduction The exact mechanisms regulating expression status of genes on the inactive X are not fully elucidated. It is known that genes escaping In female mammals, one of the two X chromosomes is inactivated inactivation are not uniformly distributed along the X chromosome to maintain equal expression between sexes by a process called X but preferentially lie in the evolutionary younger strata of the X chromosome inactivation (XCI) [1]. XCI is a complex and tightly chromosome [5,6]. It has been hypothesized that long interspersed regulated mechanism governed by the XIST gene which encodes a elements (LINE-1), which are particularly abundant on the X non-coding functional RNA coating the chromosome from which it is chromosome and are enriched in the vicinity of inactivated genes, expressed and triggers inactivation [2]. Although most genes are might control spreading of XCI [7,8]. silenced by this process, it has long been recognized that some genes Although in vitro hybrid systems have greatly contributed to our escape XCI and are expressed from both the active X (Xa) and inactive knowledge of XCI, they do not reflect the in vivo processes. In X(Xi) chromosomes. Studies using rodent-human hybrid cell lines particular, these experiments do not quantify the difference of retaining an inactive human X chromosome have estimated that expressionbetweenmalesandfemalesforgenesthatescape about 25% of X-linked genes escape inactivation to some degree [3–5]. inactivation. These genes are of primary interest since they are potential contributors to variability between sexes for phenotypic and clinical traits. Recent studies have taken benefit from the microarray Abbreviations: XCI, X Chromosome Inactivation; LCL, Lymphoblastoid Cell Line; technology to compare expression profiles of the X chromosome eQTL, Expression Quantitative Trait Locus; SNP, Single Nucleotide Polymorphism. between males and females [9–12]. The largest study, based on ⁎ Correspondingauthorat:INSERMUMRS937,FacultédeMédecinePitié- lymphoblastoid cell lines (LCLs) from 210 individuals from the four Salpêtrière, 91 bd de l'Hôpital, 75634 PARIS cedex 13, France. Fax: +33 1 40 77 97 28. HapMap populations, reported 5% of X-linked genes showing higher E-mail addresses: [email protected] (R. Castagné), [email protected] (T. Zeller), [email protected] (M. Rotival), [email protected] expression in females than in males [9], a proportion much lower than (S. Szymczak), [email protected] (V. Truong), [email protected] the 25% of genes previously thought to escape XCI [5,6]. This (A. Schillert), [email protected] (D.-A. Trégouët), [email protected] underestimation might be partly explained by an insufficient power (T. Münzel), [email protected] (A. Ziegler), [email protected] to detect small sex differences, since expression from the inactive (F. Cambien), [email protected] (S. Blankenberg), [email protected] (L. Tiret). chromosome may be low or present only in a fraction of females [5]. 1 Present address: Department of General and Interventional Cardiology, University Heart Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Here, we report the results of a large-scale study including 1467 Hamburg, Germany. population-based subjects in whom the expression profile from 0888-7543/$ – see front matter © 2011 Elsevier Inc. All rights reserved. doi:10.1016/j.ygeno.2011.06.009 R. Castagné et al. / Genomics 98 (2011) 320–326 321 circulating monocytes was assessed [13].Wefirst examined to what 2.2. Expression level of X-linked transcripts according to sex extent the mechanism of XCI was supported by this dataset including 715 X-linked transcripts. Secondly, because the impact of X-linked For each of the 715 X-linked transcripts, the median and the genetic variability on expression may have profound consequences on variance of expression levels in males were plotted against those in disease traits, as attested by numerous X-linked diseases, we females (Fig. 1). Most of the data points lay on the diagonal investigated whether cis expression quantitative loci (eQTLs) on the suggesting that XCI was globally achieved for the majority of X chromosome differed between sexes. genes. There were however a few points departing from the diagonal, most of them showing a higher expression level in femalesthaninmales. 2. Results We then focused on the transcripts that were differentially expressed between sexes using a Bonferroni-corrected significance The study was conducted in 717 women and 750 men of European threshold to account for the number of transcripts tested (7.0×10−5). origin aged 35 to 74 years that were recruited in the Gutenberg Heart We distinguished transcripts that were expressed at a higher level in Study (GHS), a community-based project conducted in a single centre females (referred to as female-biased) from those that were in Germany [13]. Genome-wide expression profiles of peripheral expressed at a higher level in males (male-biased) [14]. We observed blood monocytes were generated using Illumina HT-12 BeadChip a higher proportion of female-biased than male-biased X-linked expression arrays and genome-wide genotyping was performed using transcripts (9.37% vs 5.59%, p=0.006). We tested whether these the Affymetrix Genome-Wide Human SNP Array 6.0. proportions were different from those observed on autosomes by resampling 106 sets of 715 autosomal transcripts. There was a significant excess of female-biased transcripts on the X chromosome 2.1. X-linked transcripts detected in human monocytes compared to autosomes (9.37% vs 5.46%, pb2×10−5) whereas the difference was not significant for male-biased transcripts (5.59% vs After removing probes with a bad quality score, 1156 probes were 4.65%, p=0.10). The higher proportion of female-biased genes is in unambiguously assigned to the X chromosome, corresponding to 706 X- agreement with the fact that a fraction of X-linked genes are known or linked genes. Analyses were performed at the probe level and, for suspected to escape from silencing [5,9–12]. simplicity, the term of “transcript” was used to denote a unique probe- Because the method of monocyte enrichment used in the present hybridization product (although in few cases the same transcript could study did not yield a 100% purity and even modest heterogeneity of be targeted by several probes). We focused on transcripts whose cell content may induce artefactual associations, we checked whether expression was detected, i.e. higher than the background signal, in≥5% contamination by non-monocyte cells might affect the differences of female samples or≥5% of male samples. These represented 715 X- observed. To estimate the relative amount of RNA coming from the linked (61.8%) transcripts corresponding to 503 unique genes (Supple- different blood cell types, we applied a deconvolution algorithm mentary Table 1). The majority of X-linked transcripts were detected proposed in [15] and used cell-specific expression profiles reported in with similar detection rates in males and females (Supplementary Fig. the HaemAtlas [16] to generate surrogate variables of contamination 1). XIST (X (inactive)-specifictranscript),thegeneresponsiblefor (see Methods). Adjusting for these variables hardly modified the inactivation of one of the X copies in females, was expressed at low difference between female-biased and male-biased X-linked genes levels, yet higher than the background signal, in 62% of males. (9.51% vs 5.73%, p=0.007). Fig. 1. Comparison of expression levels of X-linked transcripts between males and females. (A) Median of expression. The pink line represents the situation where transcripts would completely escape XCI in females (log2Y=log2X+1). The only point departing from this line is XIST, the gene responsible for X inactivation of one chromosome in females. (B) Variance of expression.
Recommended publications
  • Protein Interaction Network of Alternatively Spliced Isoforms from Brain Links Genetic Risk Factors for Autism
    ARTICLE Received 24 Aug 2013 | Accepted 14 Mar 2014 | Published 11 Apr 2014 DOI: 10.1038/ncomms4650 OPEN Protein interaction network of alternatively spliced isoforms from brain links genetic risk factors for autism Roser Corominas1,*, Xinping Yang2,3,*, Guan Ning Lin1,*, Shuli Kang1,*, Yun Shen2,3, Lila Ghamsari2,3,w, Martin Broly2,3, Maria Rodriguez2,3, Stanley Tam2,3, Shelly A. Trigg2,3,w, Changyu Fan2,3, Song Yi2,3, Murat Tasan4, Irma Lemmens5, Xingyan Kuang6, Nan Zhao6, Dheeraj Malhotra7, Jacob J. Michaelson7,w, Vladimir Vacic8, Michael A. Calderwood2,3, Frederick P. Roth2,3,4, Jan Tavernier5, Steve Horvath9, Kourosh Salehi-Ashtiani2,3,w, Dmitry Korkin6, Jonathan Sebat7, David E. Hill2,3, Tong Hao2,3, Marc Vidal2,3 & Lilia M. Iakoucheva1 Increased risk for autism spectrum disorders (ASD) is attributed to hundreds of genetic loci. The convergence of ASD variants have been investigated using various approaches, including protein interactions extracted from the published literature. However, these datasets are frequently incomplete, carry biases and are limited to interactions of a single splicing isoform, which may not be expressed in the disease-relevant tissue. Here we introduce a new interactome mapping approach by experimentally identifying interactions between brain-expressed alternatively spliced variants of ASD risk factors. The Autism Spliceform Interaction Network reveals that almost half of the detected interactions and about 30% of the newly identified interacting partners represent contribution from splicing variants, emphasizing the importance of isoform networks. Isoform interactions greatly contribute to establishing direct physical connections between proteins from the de novo autism CNVs. Our findings demonstrate the critical role of spliceform networks for translating genetic knowledge into a better understanding of human diseases.
    [Show full text]
  • Contiguous Deletion of the NDP, MAOA, MAOB, and EFHC2 Genes in a Patient with Norrie Disease, Severe Psychomotor Retardation and Myoclonic Epilepsy
    ß 2007 Wiley-Liss, Inc. American Journal of Medical Genetics Part A 143A:916–920 (2007) Contiguous Deletion of the NDP, MAOA, MAOB, and EFHC2 Genes in a Patient With Norrie Disease, Severe Psychomotor Retardation and Myoclonic Epilepsy L. Rodriguez-Revenga,1,2 I. Madrigal,1,2 L.S. Alkhalidi,3 L. Armengol,4 E. Gonza´lez,4 C. Badenas,1,2 X. Estivill,1,5 and M. Mila`1,2* 1Biochemistry and Molecular Genetics Department, Hospital Clı´nic, Barcelona, Spain 2IDIBAPS (Institut d’Investigacions Biome`diques August Pi i Sunyer), Barcelona, Spain 3Department of Health and Medical Services, Rashid Hospital, Dubai, United Arab Emirates 4Genes and Disease Programme, Centre for Genomic Regulation (CRG), Barcelona Biomedical Research Park, Barcelona, Spain 5Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain Received 20 February 2006; Accepted 7 September 2006 Norrie disease (ND) is an X-linked disorder, inherited as a Clinical features of the proband include bilateral retinal recessive trait that, therefore, mostly affects males. The gene detachment, microcephaly, severe psychomotor retardation responsible for ND, called NDP, maps to the short arm of without verbal language skills acquired, and epilepsy. The chromosome X (Xp11.4-p11.3). We report here an atypical identification and molecular characterization of this case case of ND, consisting of a patient harboring a large reinforces the idea of a new contiguous gene syndrome that submicroscopic deletion affecting not only the NDP gene would explain the complex phenotype shared by atypical but also the MAOA, MAOB, and EFHC2 genes. Microarray ND patients. ß 2007 Wiley-Liss, Inc.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Protein Localization Screening in Vivo Reveals Novel Regulators of Multiciliated Cell Development and Function Fan Tu1, Jakub Sedzinski1,2, Yun Ma1,3, Edward M
    © 2018. Published by The Company of Biologists Ltd | Journal of Cell Science (2018) 131, jcs206565. doi:10.1242/jcs.206565 SHORT REPORT Protein localization screening in vivo reveals novel regulators of multiciliated cell development and function Fan Tu1, Jakub Sedzinski1,2, Yun Ma1,3, Edward M. Marcotte1 and John B. Wallingford1,* ABSTRACT RESULTS AND DISCUSSION Multiciliated cells (MCCs) drive fluid flow in diverse tubular organs High-content screening of protein localization in MCCs in vivo and are essential for the development and homeostasis of the vertebrate central nervous system, airway and reproductive tracts. High-content screening of protein localization is a powerful These cells are characterized by dozens or hundreds of motile cilia approach for linking genomics to cell biology (Boutros et al., Xenopus that beat in a coordinated and polarized manner. In recent years, 2015), so we turned to embryos, where the epidermis genomic studies have not only elucidated the transcriptional develops as a mix of MCCs and mucus-secreting cells similar to in hierarchy for MCC specification but also identified myriad new the mammalian airway (Hayes et al., 2007; Walentek and Quigley, proteins that govern MCC ciliogenesis, cilia beating and cilia 2017; Werner and Mitchell, 2011). Molecular mechanisms of MCC Xenopus polarization. Interestingly, this burst of genomic data has also development and function are highly conserved between highlighted that proteins with no obvious role in cilia do, in fact, have and mammals, including humans (e.g. Boon et al., 2014; Toriyama important ciliary functions. Understanding the function of proteins et al., 2016; Wallmeier et al., 2014, 2016; Zariwala et al., 2013), and Xenopus with little prior history of study presents a special challenge, MCCs are very large and present on the external surface of especially when faced with large numbers of such proteins.
    [Show full text]
  • Genome-Wide Meta-Analysis Identifies 127 Open-Angle Glaucoma Loci with Consistent Effect Across Ancestries
    UC San Diego UC San Diego Previously Published Works Title Genome-wide meta-analysis identifies 127 open-angle glaucoma loci with consistent effect across ancestries. Permalink https://escholarship.org/uc/item/6fn1m7tr Journal Nature communications, 12(1) ISSN 2041-1723 Authors Gharahkhani, Puya Jorgenson, Eric Hysi, Pirro et al. Publication Date 2021-02-24 DOI 10.1038/s41467-020-20851-4 Peer reviewed eScholarship.org Powered by the California Digital Library University of California ARTICLE https://doi.org/10.1038/s41467-020-20851-4 OPEN Genome-wide meta-analysis identifies 127 open-angle glaucoma loci with consistent effect across ancestries ✉ Puya Gharahkhani 1,190 , Eric Jorgenson 2,190, Pirro Hysi 3,190, Anthony P. Khawaja 4,5,190, Sarah Pendergrass6,190, Xikun Han 1, Jue Sheng Ong 1, Alex W. Hewitt 7,8, Ayellet V. Segrè9, John M. Rouhana 9, Andrew R. Hamel9, Robert P. Igo Jr 10, Helene Choquet 2, Ayub Qassim11, Navya S. Josyula 12, Jessica N. Cooke Bailey 10,13, Pieter W. M. Bonnemaijer 14,15,16, Adriana Iglesias 14,15,17, Owen M. Siggs 11, Terri L. Young 18, Veronique Vitart 19, 1234567890():,; Alberta A. H. J. Thiadens 14,15, Juha Karjalainen20,21,22, Steffen Uebe23, Ronald B. Melles24, K. Saidas Nair25, Robert Luben 5, Mark Simcoe 3,26,27, Nishani Amersinghe28, Angela J. Cree 29, Rene Hohn30,31, Alicia Poplawski 32, Li Jia Chen33, Shi-Song Rong 9,33, Tin Aung34,35,36, Eranga Nishanthie Vithana 34,37, NEIGHBORHOOD consortium*, ANZRAG consortium*, Biobank Japan project*, FinnGen study*, UK Biobank Eye and Vision Consortium*, GIGA study group*, 23 and Me Research Team*, Gen Tamiya38,39, Yukihiro Shiga40, Masayuki Yamamoto 38, Toru Nakazawa40,41,42,43, Hannah Currant 44, Ewan Birney 44, Xin Wang 45, Adam Auton45, Michelle K.
    [Show full text]
  • Evolutionary Proteomics Uncovers Ciliary Signaling Components 2 3 Monika Abedin Sigg1, Tabea Menchen2, Jeffery Johnson3, Chanjae Lee4, Semil P
    bioRxiv preprint doi: https://doi.org/10.1101/153437; this version posted June 22, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Evolutionary proteomics uncovers ciliary signaling components 2 3 Monika Abedin Sigg1, Tabea Menchen2, Jeffery Johnson3, Chanjae Lee4, Semil P. Choksi1, Galo 4 Garcia 3rd,1, Henriette Busengdal5, Gerard Dougherty2, Petra Pennekamp2, Claudius Werner2, 5 Fabian Rentzsch5, Nevan Krogan3,6, John B. Wallingford4, Heymut Omran2 and Jeremy F. 6 Reiter1* 7 8 Affiliations 9 1Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of 10 California, San Francisco, CA 94158, USA 11 2Department of General Pediatrics, University of Muenster, Muenster 48149, Germany 12 3Gladstone Institute of Cardiovascular Disease and Gladstone Institute of Virology and 13 Immunology, San Francisco, CA 94158, USA 14 4Department of Molecular Biosciences, Center for Systems and Synthetic Biology and Institute 15 for Cellular and Molecular Biology, University of Texas at Austin, Austin 78712, Texas 16 5Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen 5008, 17 Norway 18 6Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 19 CA 94158, USA 20 *Correspondence: [email protected] 21 22 1 bioRxiv preprint doi: https://doi.org/10.1101/153437; this version posted June 22, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 23 ABSTRACT 24 Cilia are organelles specialized for movement and signaling. To infer when during animal 25 evolution signaling pathways became associated with cilia, we characterized the proteomes of 26 cilia from three organisms: sea urchins, sea anemones and choanoflagellates.
    [Show full text]
  • Genomic Approach in Idiopathic Intellectual Disability Maria De Fátima E Costa Torres
    ESTUDOS DE 8 01 PDPGM 2 CICLO Genomic approach in idiopathic intellectual disability Maria de Fátima e Costa Torres D Autor. Maria de Fátima e Costa Torres D.ICBAS 2018 Genomic approach in idiopathic intellectual disability Genomic approach in idiopathic intellectual disability Maria de Fátima e Costa Torres SEDE ADMINISTRATIVA INSTITUTO DE CIÊNCIAS BIOMÉDICAS ABEL SALAZAR FACULDADE DE MEDICINA MARIA DE FÁTIMA E COSTA TORRES GENOMIC APPROACH IN IDIOPATHIC INTELLECTUAL DISABILITY Tese de Candidatura ao grau de Doutor em Patologia e Genética Molecular, submetida ao Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto Orientadora – Doutora Patrícia Espinheira de Sá Maciel Categoria – Professora Associada Afiliação – Escola de Medicina e Ciências da Saúde da Universidade do Minho Coorientadora – Doutora Maria da Purificação Valenzuela Sampaio Tavares Categoria – Professora Catedrática Afiliação – Faculdade de Medicina Dentária da Universidade do Porto Coorientadora – Doutora Filipa Abreu Gomes de Carvalho Categoria – Professora Auxiliar com Agregação Afiliação – Faculdade de Medicina da Universidade do Porto DECLARAÇÃO Dissertação/Tese Identificação do autor Nome completo _Maria de Fátima e Costa Torres_ N.º de identificação civil _07718822 N.º de estudante __ 198600524___ Email institucional [email protected] OU: [email protected] _ Email alternativo [email protected] _ Tlf/Tlm _918197020_ Ciclo de estudos (Mestrado/Doutoramento) _Patologia e Genética Molecular__ Faculdade/Instituto _Instituto de Ciências
    [Show full text]
  • Mouse Efhc2 Conditional Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Efhc2 Conditional Knockout Project (CRISPR/Cas9) Objective: To create a Efhc2 conditional knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Efhc2 gene (NCBI Reference Sequence: NM_028916 ; Ensembl: ENSMUSG00000025038 ) is located on Mouse chromosome X. 15 exons are identified, with the ATG start codon in exon 1 and the TGA stop codon in exon 15 (Transcript: ENSMUST00000026014). Exon 3 will be selected as conditional knockout region (cKO region). Deletion of this region should result in the loss of function of the Mouse Efhc2 gene. To engineer the targeting vector, homologous arms and cKO region will be generated by PCR using BAC clone RP24-299C2 as template. Cas9, gRNA and targeting vector will be co-injected into fertilized eggs for cKO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Exon 3 starts from about 10.31% of the coding region. The knockout of Exon 3 will result in frameshift of the gene. The size of intron 2 for 5'-loxP site insertion: 37900 bp, and the size of intron 3 for 3'-loxP site insertion: 12702 bp. The size of effective cKO region: ~651 bp. The cKO region does not have any other known gene. Page 1 of 7 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele gRNA region 5' gRNA region 3' 1 3 15 Targeting vector Targeted allele Constitutive KO allele (After Cre recombination) Legends Exon of mouse Efhc2 Homology arm cKO region loxP site Page 2 of 7 https://www.alphaknockout.com Overview of the Dot Plot Window size: 10 bp Forward Reverse Complement Sequence 12 Note: The sequence of homologous arms and cKO region is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • Genetic Susceptibility in Juvenile Myoclonic Epilepsy: Systematic Review of Genetic Association Studies
    RESEARCH ARTICLE Genetic susceptibility in Juvenile Myoclonic Epilepsy: Systematic review of genetic association studies Bruna Priscila dos Santos1, Chiara Rachel Maciel Marinho1, Thalita Ewellyn Batista Sales Marques1, Layanne Kelly Gomes Angelo1, MaõÂsa Vieira da Silva Malta1, Marcelo Duzzioni2, Olagide Wagner de Castro3, João Pereira Leite4, Fabiano Timbo Barbosa5, Daniel Leite Go es GitaõÂ1* a1111111111 1 Department of Cellular and Molecular Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, Maceio, Alagoas, Brazil, 2 Department of Pharmacology, Institute of Biological a1111111111 Sciences and Health, Federal University of Alagoas, Maceio, Alagoas, Brazil, 3 Department of Physiology, a1111111111 Institute of Biological Sciences and Health, Federal University of Alagoas, Maceio, Alagoas, Brazil, 4 Division a1111111111 of Neurology, Department of Neurosciences and Behavioral Sciences, Ribeirão Preto School of Medicine, a1111111111 University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 5 School of Medicine, Federal University of Alagoas, Maceio, Alagoas, Brazil * [email protected] OPEN ACCESS Citation: Santos BPd, Marinho CRM, Marques Abstract TEBS, Angelo LKG, Malta MVdS, Duzzioni M, et al. (2017) Genetic susceptibility in Juvenile Myoclonic Epilepsy: Systematic review of genetic association Background studies. PLoS ONE 12(6): e0179629. https://doi. Several genetic association investigations have been performed over the last three decades org/10.1371/journal.pone.0179629 to identify variants underlying Juvenile Myoclonic Epilepsy (JME). Here, we evaluate the Editor: Klaus Brusgaard, Odense University accumulating findings and provide an updated perspective of these studies. Hospital, DENMARK Received: September 9, 2016 Methodology Accepted: June 1, 2017 A systematic literature search was conducted using the PubMed, Embase, Scopus, Lilacs, Published: June 21, 2017 epiGAD, Google Scholar and Sigle up to February 12, 2016.
    [Show full text]
  • Juvenile Myoclonic Epilepsy Is Not Associated with the DRPLA Gene in a European Population
    in vivo 28: 1193-1196 (2014) Juvenile Myoclonic Epilepsy Is Not Associated with the DRPLA Gene in a European Population CHRISTOS YAPIJAKIS1,2, STERGIOS GATZONIS3, SOTIRIOS YOUROUKOS4, VASILIKI KOLLIA2, STELLA KARACHRISTIANOU5 and MARIA ANAGNOSTOULI1 1First Department of Neurology, Eginition Hospital, University of Athens Medical School, Athens, Greece; 2Cephalogenetics Research Center, Athens, Greece; 3Department of Neurosurgery, University of Athens Medical School, Evangelismos Hospital, Athens, Greece; 4First Department of Pediatrics, Agia Sophia Hospital, University of Athens Medical School, Athens, Greece; 5Third Department of Neurology, Papanikolaou Hospital, Aristotelian University of Thessaloniki Medical School, Thessaloniki, Greece Abstract. Background: Juvenile myoclonic epilepsy (JME), of generalized epilepsy (2-4). It is distinguished from other is an early-onset inherited generalized epilepsy which generalized epilepsies by its onset in early adolescence and displays genetic heterogeneity, with at least 10 known loci. the observation of myoclonic jerks, which are not necessarily Another neurogenetic disease, dentato-rubro-pallido-luysian a prelude to a major seizure and usually occur in the atrophy (DRPLA) presents three clinical phenotypes, one of morning, upon awakening (3, 5). The presentation of all JME which in Japanese displays many similarities to JME. Aim: cases seems to belong in a defined clinical spectrum The purpose of this study was to investigate whether the according to the diagnostic criteria of the International DRPLA gene is associated with JME in Caucasians. League Against Epilepsy (6, 7). Patients and Methods: The CAG repeat polymorphism in the Nevertheless, JME displays genetic heterogeneity, with at DRPLA gene, which is expanded in patients with DRPLA, least 10 loci identified so far, on chromosomes 6p21, 15q14, was examined with polymerase chain reaction amplification 2q23.3, 5q34, 1p36.33, Xp11.3, 5q12-q14, 6p12.2, 3q27.1, in 107 individuals of Greek origin, including 24 patients with and 2q33-q36 (8-17).
    [Show full text]
  • A Genome Model Linking Birth Defects to Infections
    bioRxiv preprint doi: https://doi.org/10.1101/674093; this version posted June 19, 2019. 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-ND 4.0 International license. A Genome Model Linking Birth Defects to Infections Bernard Friedenson Deparment of Biochemistry and Molecular Genetics College of Medicine University of Illinois Chicago Chicago, Illinois 60612 Abstract The purpose of this study was to test the hypothesis that infections are linked to chromosomal anomalies that cause neurodevelopmental disorders. In chil- dren with disorders in the development of their nervous systems, chromosome anomalies known to cause these disorders were compared to microbial DNA, including known teratogens. Genes essential for neurons, lymphatic drainage, immunity, circulation, angiogenesis, cell barriers, structure, epigenetic and chro- matin modifications were all found close together in polyfunctional clusters that were deleted or rearranged in neurodevelopmental disorders. In some patients, epigenetic driver mutations also changed access to large chromosome segments. These changes account for immune, circulatory, and structural deficits that ac- company neurologic deficits. Specific and repetitive human DNA encompassing large deletions matched infections and passed rigorous artifact tests. Deletions of up to millions of bases accompanied infection-matching sequences and caused massive changes in the homologies to foreign DNAs. In data from three inde- pendent studies of private, familial and recurrent chromosomal rearrangements, massive changes in homologous microbiomes were found and may drive rear- rangements and encourage pathogens. At least one chromosomal anomaly was found to consist of human DNA fragments with a gap that corresponded to a piece of integrated foreign DNA.
    [Show full text]
  • Introgression of Regulatory Alleles and a Missense Coding Mutation Drive Plumage Pattern Diversity in the Rock Pigeon
    RESEARCH ARTICLE Introgression of regulatory alleles and a missense coding mutation drive plumage pattern diversity in the rock pigeon Anna I Vickrey1, Rebecca Bruders1, Zev Kronenberg2†, Emma Mackey1‡, Ryan J Bohlender3, Emily T Maclary1, Raquel Maynez1, Edward J Osborne2, Kevin P Johnson4, Chad D Huff3, Mark Yandell2, Michael D Shapiro1* 1School of Biological Sciences, University of Utah, Salt Lake City, United States; 2Department of Human Genetics, University of Utah, Salt Lake City, United States; 3Department of Epidemiology, MD Anderson Cancer Center, University of Texas, Houston, United States; 4Illinois Natural History Survey, Prairie Research Institute, University of Illinois Urbana-Champaign, Champaign, United States Abstract Birds and other vertebrates display stunning variation in pigmentation patterning, yet the genes controlling this diversity remain largely unknown. Rock pigeons (Columba livia) are fundamentally one of four color pattern phenotypes, in decreasing order of melanism: T-check, checker, bar (ancestral), or barless. Using whole-genome scans, we identified NDP as a candidate gene for this variation. Allele-specific expression differences in NDP indicate cis-regulatory divergence between ancestral and melanistic alleles. Sequence comparisons suggest that derived *For correspondence: alleles originated in the speckled pigeon (Columba guinea), providing a striking example of [email protected] introgression. In contrast, barless rock pigeons have an increased incidence of vision defects and, like human families with hereditary blindness, carry start-codon mutations in NDP. In summary, we Present address: †Phase find that both coding and regulatory variation in the same gene drives wing pattern diversity, and Genomics, Seattle, United States; ‡Department of post-domestication introgression supplied potentially advantageous melanistic alleles to feral Molecular and Cell Biology, populations of this ubiquitous urban bird.
    [Show full text]