Microrna Dysregulation in Neuropsychiatric Disorders and Cognitive Dysfunction

Total Page:16

File Type:pdf, Size:1020Kb

Microrna Dysregulation in Neuropsychiatric Disorders and Cognitive Dysfunction MicroRNA Dysregulation in Neuropsychiatric Disorders and Cognitive Dysfunction Pei-Ken Hsu Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy under the Executive Committee of the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2012 © 2012 Pei-Ken Hsu All rights reserved ABSTRACT MicroRNA Dysregulation in Neuropsychiatric Disorders and Cognitive Dysfunction Pei-Ken Hsu MicroRNAs (miRNAs) are evolutionarily-conserved small non-coding RNAs that are important posttranscriptional regulators of gene expression. Genetic Variants may cause microRNA dysregulation and the concomitant aberrant target expression. The dysregulation of one or a few targets may in turn lead to functional consequences ranging from phenotypic variations to disease conditions. In this thesis, I present our studies of mouse models of two human genetic variants − a rare copy number variant (CNV), 22q11.2 microdeletions, and a common single nucleotide polymorphism (SNP), BDNF Val66Met . 22q11.2 microdeletions result in specific cognitive deficits and high risk to develop schizophrenia. Analysis of Df(16)A +/– mice, which model this microdeletion, revealed abnormalities in the formation of neuronal dendrites and spines as well as microRNA dysregulation in brain. We show a drastic reduction of miR- 185, which resides within the 22q11.2 locus, to levels more than expected by a hemizygous deletion and demonstrate that this reduction impairs dendritic and spine development. miR-185 targets and represses, through an evolutionary conserved target site, a previously unknown inhibitor of these processes that resides in the Golgi apparatus. Sustained derepression of this inhibitor after birth represents the most robust transcriptional disturbance in the brains of Df(16)A +/− mice and could affect the formation and maintenance of neural circuits. Reduction of miR-185 also has milder effects on the expression of a group of Golgi-related genes. One the other hand, BNDF Val66Met results in impaired activity-dependent secretion of BDNF from neuronal terminals and affects episodic memory and affective behaviors. We found a modest reduction of miR-146b which causes derepression of mRNA and/or protein levels of a few targets. Our findings add to the growing evidence of the pivotal involvement of miRNAs in the development of neuropsychiatric disorders and cognitive dysfunction. In addition, the identification of key players in miRNA dysregulation has implications for both basic and translational research in psychiatric disorders and cognitive dysfunction. Table of Contents Chapter I − Tiny Regulators with Profound Impact in Neuropsychiatric Disorders ix 1.1 MicroRNAs-mediated Regulation ..................................................................................................... 1 1.2 Altered microRNA Expression and Function in Neuropsychiatric Disorders ............................. 3 1.2.1 Schizophrenia .............................................................................................................................. 3 1.2.2 Autism Spectrum Disorders ......................................................................................................... 8 1.2.3 Rett Syndrome ........................................................................................................................... 10 1.2.4 Fragile X Syndrome ................................................................................................................... 12 1.2.5 Tourette's Syndrome .................................................................................................................. 13 1.2.6 Down Syndrome ........................................................................................................................ 14 1.3 Potential Mechanistic Connections between microRNA Dysregulation and Neuropsychiatric Disorders ........................................................................................................... 15 1.3.1 Insights from Global Disruption of miRNA Biogenesis and Action ............................................ 15 1.3.2 Individual miRNAs Modulate Dendritic Complexity and Spine Morphology in Neurons ............ 16 1.3.3 Individual miRNAs Modulate Neurogenesis, Neuronal Proliferation, Migration and Integration ................................................................................................................................. 18 1.3.4 Individual miRNAs Modulate Neuronal Electrophysiological Properties in Response to Neuronal Activity ....................................................................................................................... 20 1.4 Summary ........................................................................................................................................... 20 1.5 References ........................................................................................................................................ 24 Chapter II − A Major Downstream Effector of MicroRNA Dysregulation in 22q11.2 Genomic Losses ...................................................................... 36 2.1 Introduction ...................................................................................................................................... 36 2.1.1 22q11.2 Microdeletions and Schizophrenia ............................................................................... 36 2.1.2 A Mouse Model of 22q11.2 Microdeletion ................................................................................. 37 2.1.3 miRNA Dysregulation in of 22q11.2 Microdeletion Mouse Model ............................................. 39 2.1.4 In this Chapter ............................................................................................................................ 41 i 2.2 Results .............................................................................................................................................. 41 2.2.1 A Drastic Reduction of miR-185 Levels in Df(16)A +/– Mice ........................................................ 41 2.2.2 A Primary Transcriptional Consequence of 22q11.2 Genomic Losses ..................................... 43 2.2.3 2310044H10Rik as a Major Downstream Target of miRNAs Dysregulated in Df(16)A +/– Mice .......................................................................................................................... 45 2.3 Discussion ........................................................................................................................................ 51 2.3.1 CNV-associated miRNA Dysregulation ..................................................................................... 51 2.3.2 Convergent Downregulation of miRNAs in Schizophrenia Patients and Df(16)A +/– Mice .......... 53 2.3.3 Impact of Modest Dysregulation of miRNAs .............................................................................. 54 2.3.4 Not all miRNA Targets are Created Equal? ............................................................................... 55 2.4 Summary ........................................................................................................................................... 56 2.5 Methods ............................................................................................................................................ 63 2.6 References ........................................................................................................................................ 69 Chapter III − Dysregulation of A Novel Inhibitor of Dendritic and Spine Morphogenesis In Df(16)A +/– Mice ........................................................ 75 3.1 Introduction ...................................................................................................................................... 75 3.1.1 miRNA Regulation of Dendritic Arborization .............................................................................. 76 3.1.2 miRNA Regulation of Spine Morphogenesis ............................................................................. 77 3.1.3 Structural Alterations in Df(16)A +/– Neurons ............................................................................. 79 3.1.4 In this Chapter ............................................................................................................................ 80 3.2 Results .............................................................................................................................................. 81 3.2.1 Generation of Mirta22 Specific Antibody ................................................................................... 81 3.2.2 Mirta22 is a Neuronal Protein Residing in the Golgi Apparatus ................................................ 81 3.2.3 Coordinated Mild Dysregulation of Golgi-related Genes due to miR-185 Reduction ................ 85 3.2.4 Altered miR-185 Levels Contribute to Structural Alterations of Df(16)A +/− Neurons .................. 86 3.2.5 Elevation of Mirta22 Levels Inhibits Dendritic and Spine Development in Df(16)A +/– Neurons .................................................................................................................... 89 ii 3.3 Discussion ........................................................................................................................................ 92 3.3.1 What do We Know about Mirta22 Protein? ................................................................................ 92 3.3.2 A Neuronal Inhibitor
Recommended publications
  • Seq2pathway Vignette
    seq2pathway Vignette Bin Wang, Xinan Holly Yang, Arjun Kinstlick May 19, 2021 Contents 1 Abstract 1 2 Package Installation 2 3 runseq2pathway 2 4 Two main functions 3 4.1 seq2gene . .3 4.1.1 seq2gene flowchart . .3 4.1.2 runseq2gene inputs/parameters . .5 4.1.3 runseq2gene outputs . .8 4.2 gene2pathway . 10 4.2.1 gene2pathway flowchart . 11 4.2.2 gene2pathway test inputs/parameters . 11 4.2.3 gene2pathway test outputs . 12 5 Examples 13 5.1 ChIP-seq data analysis . 13 5.1.1 Map ChIP-seq enriched peaks to genes using runseq2gene .................... 13 5.1.2 Discover enriched GO terms using gene2pathway_test with gene scores . 15 5.1.3 Discover enriched GO terms using Fisher's Exact test without gene scores . 17 5.1.4 Add description for genes . 20 5.2 RNA-seq data analysis . 20 6 R environment session 23 1 Abstract Seq2pathway is a novel computational tool to analyze functional gene-sets (including signaling pathways) using variable next-generation sequencing data[1]. Integral to this tool are the \seq2gene" and \gene2pathway" components in series that infer a quantitative pathway-level profile for each sample. The seq2gene function assigns phenotype-associated significance of genomic regions to gene-level scores, where the significance could be p-values of SNPs or point mutations, protein-binding affinity, or transcriptional expression level. The seq2gene function has the feasibility to assign non-exon regions to a range of neighboring genes besides the nearest one, thus facilitating the study of functional non-coding elements[2]. Then the gene2pathway summarizes gene-level measurements to pathway-level scores, comparing the quantity of significance for gene members within a pathway with those outside a pathway.
    [Show full text]
  • Investigating the Genetic Basis of Cisplatin-Induced Ototoxicity in Adult South African Patients
    --------------------------------------------------------------------------- Investigating the genetic basis of cisplatin-induced ototoxicity in adult South African patients --------------------------------------------------------------------------- by Timothy Francis Spracklen SPRTIM002 SUBMITTED TO THE UNIVERSITY OF CAPE TOWN In fulfilment of the requirements for the degree MSc(Med) Faculty of Health Sciences UNIVERSITY OF CAPE TOWN University18 December of Cape 2015 Town Supervisor: Prof. Rajkumar S Ramesar Co-supervisor: Ms A Alvera Vorster Division of Human Genetics, Department of Pathology, University of Cape Town 1 The copyright of this thesis vests in the author. No quotation from it or information derived from it is to be published without full acknowledgement of the source. The thesis is to be used for private study or non- commercial research purposes only. Published by the University of Cape Town (UCT) in terms of the non-exclusive license granted to UCT by the author. University of Cape Town Declaration I, Timothy Spracklen, hereby declare that the work on which this dissertation/thesis is based is my original work (except where acknowledgements indicate otherwise) and that neither the whole work nor any part of it has been, is being, or is to be submitted for another degree in this or any other university. I empower the university to reproduce for the purpose of research either the whole or any portion of the contents in any manner whatsoever. Signature: Date: 18 December 2015 ' 2 Contents Abbreviations ………………………………………………………………………………….. 1 List of figures …………………………………………………………………………………... 6 List of tables ………………………………………………………………………………….... 7 Abstract ………………………………………………………………………………………… 10 1. Introduction …………………………………………………………………………………. 11 1.1 Cancer …………………………………………………………………………….. 11 1.2 Adverse drug reactions ………………………………………………………….. 12 1.3 Cisplatin …………………………………………………………………………… 12 1.3.1 Cisplatin’s mechanism of action ……………………………………………… 13 1.3.2 Adverse reactions to cisplatin therapy ……………………………………….
    [Show full text]
  • (P -Value<0.05, Fold Change≥1.4), 4 Vs. 0 Gy Irradiation
    Table S1: Significant differentially expressed genes (P -Value<0.05, Fold Change≥1.4), 4 vs. 0 Gy irradiation Genbank Fold Change P -Value Gene Symbol Description Accession Q9F8M7_CARHY (Q9F8M7) DTDP-glucose 4,6-dehydratase (Fragment), partial (9%) 6.70 0.017399678 THC2699065 [THC2719287] 5.53 0.003379195 BC013657 BC013657 Homo sapiens cDNA clone IMAGE:4152983, partial cds. [BC013657] 5.10 0.024641735 THC2750781 Ciliary dynein heavy chain 5 (Axonemal beta dynein heavy chain 5) (HL1). 4.07 0.04353262 DNAH5 [Source:Uniprot/SWISSPROT;Acc:Q8TE73] [ENST00000382416] 3.81 0.002855909 NM_145263 SPATA18 Homo sapiens spermatogenesis associated 18 homolog (rat) (SPATA18), mRNA [NM_145263] AA418814 zw01a02.s1 Soares_NhHMPu_S1 Homo sapiens cDNA clone IMAGE:767978 3', 3.69 0.03203913 AA418814 AA418814 mRNA sequence [AA418814] AL356953 leucine-rich repeat-containing G protein-coupled receptor 6 {Homo sapiens} (exp=0; 3.63 0.0277936 THC2705989 wgp=1; cg=0), partial (4%) [THC2752981] AA484677 ne64a07.s1 NCI_CGAP_Alv1 Homo sapiens cDNA clone IMAGE:909012, mRNA 3.63 0.027098073 AA484677 AA484677 sequence [AA484677] oe06h09.s1 NCI_CGAP_Ov2 Homo sapiens cDNA clone IMAGE:1385153, mRNA sequence 3.48 0.04468495 AA837799 AA837799 [AA837799] Homo sapiens hypothetical protein LOC340109, mRNA (cDNA clone IMAGE:5578073), partial 3.27 0.031178378 BC039509 LOC643401 cds. [BC039509] Homo sapiens Fas (TNF receptor superfamily, member 6) (FAS), transcript variant 1, mRNA 3.24 0.022156298 NM_000043 FAS [NM_000043] 3.20 0.021043295 A_32_P125056 BF803942 CM2-CI0135-021100-477-g08 CI0135 Homo sapiens cDNA, mRNA sequence 3.04 0.043389246 BF803942 BF803942 [BF803942] 3.03 0.002430239 NM_015920 RPS27L Homo sapiens ribosomal protein S27-like (RPS27L), mRNA [NM_015920] Homo sapiens tumor necrosis factor receptor superfamily, member 10c, decoy without an 2.98 0.021202829 NM_003841 TNFRSF10C intracellular domain (TNFRSF10C), mRNA [NM_003841] 2.97 0.03243901 AB002384 C6orf32 Homo sapiens mRNA for KIAA0386 gene, partial cds.
    [Show full text]
  • DGCR6 at the Proximal Part of the Digeorge Critical Region Is Involved in Conotruncal Heart Defects
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Institutional Repository : the EHIME area OPEN Citation: Human Genome Variation (2015) 2, 15004; doi:10.1038/hgv.2015.4 © 2015 The Japan Society of Human Genetics All rights reserved 2054-345X/15 www.nature.com/hgv ARTICLE DGCR6 at the proximal part of the DiGeorge critical region is involved in conotruncal heart defects Wenming Gao1, Takashi Higaki1, Minenori Eguchi-Ishimae1, Hidehiko Iwabuki1, Zhouying Wu1, Eiichi Yamamoto2, Hidemi Takata1, Masaaki Ohta1, Issei Imoto3, Eiichi Ishii1 and Mariko Eguchi1 Cardiac anomaly is one of the hallmarks of DiGeorge syndrome (DGS), observed in approximately 80% of patients. It often shows a characteristic morphology, termed as conotruncal heart defects. In many cases showing only the conotruncal heart defect, deletion of 22q11.2 region cannot be detected by fluorescence in situ hybridization (FISH), which is used to detect deletion in DGS. We investigated the presence of genomic aberrations in six patients with congenital conotruncal heart defects, who show no deletion at 22q11.2 in an initial screening by FISH. In these patients, no abnormalities were identified in the coding region of the TBX1 gene, one of the key genes responsible for the phenotype of DGS. However, when copy number alteration was analyzed by high-resolution array analysis, a small deletion or duplication in the proximal end of DiGeorge critical region was detected in two patients. The affected region contains the DGCR6 and PRODH genes. DGCR6 has been reported to affect the expression of the TBX1 gene. Our results suggest that altered dosage of gene(s) other than TBX1, possibly DGCR6, may also be responsible for the development of conotruncal heart defects observed in patients with DGS and, in particular, in those with stand-alone conotruncal heart defects.
    [Show full text]
  • Human Lectins, Their Carbohydrate Affinities and Where to Find Them
    biomolecules Review Human Lectins, Their Carbohydrate Affinities and Where to Review HumanFind Them Lectins, Their Carbohydrate Affinities and Where to FindCláudia ThemD. Raposo 1,*, André B. Canelas 2 and M. Teresa Barros 1 1, 2 1 Cláudia D. Raposo * , Andr1 é LAQVB. Canelas‐Requimte,and Department M. Teresa of Chemistry, Barros NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829‐516 Caparica, Portugal; [email protected] 12 GlanbiaLAQV-Requimte,‐AgriChemWhey, Department Lisheen of Chemistry, Mine, Killoran, NOVA Moyne, School E41 of ScienceR622 Co. and Tipperary, Technology, Ireland; canelas‐ [email protected] NOVA de Lisboa, 2829-516 Caparica, Portugal; [email protected] 2* Correspondence:Glanbia-AgriChemWhey, [email protected]; Lisheen Mine, Tel.: Killoran, +351‐212948550 Moyne, E41 R622 Tipperary, Ireland; [email protected] * Correspondence: [email protected]; Tel.: +351-212948550 Abstract: Lectins are a class of proteins responsible for several biological roles such as cell‐cell in‐ Abstract:teractions,Lectins signaling are pathways, a class of and proteins several responsible innate immune for several responses biological against roles pathogens. such as Since cell-cell lec‐ interactions,tins are able signalingto bind to pathways, carbohydrates, and several they can innate be a immuneviable target responses for targeted against drug pathogens. delivery Since sys‐ lectinstems. In are fact, able several to bind lectins to carbohydrates, were approved they by canFood be and a viable Drug targetAdministration for targeted for drugthat purpose. delivery systems.Information In fact, about several specific lectins carbohydrate were approved recognition by Food by andlectin Drug receptors Administration was gathered for that herein, purpose. plus Informationthe specific organs about specific where those carbohydrate lectins can recognition be found by within lectin the receptors human was body.
    [Show full text]
  • Systematic Analysis of Palatal Transcriptome to Identify Cleft Palate Genes Within Tgfβ3-Knockout Mice Alleles: RNA-Seq Analysis of Tgfβ3 Mice Ozturk Et Al
    Systematic analysis of palatal transcriptome to identify cleft palate genes within TGFβ3-knockout mice alleles: RNA-Seq analysis of TGFβ3 Mice Ozturk et al. Ozturk et al. BMC Genomics 2013, 14:113 http://www.biomedcentral.com/1471-2164/14/113 Ozturk et al. BMC Genomics 2013, 14:113 http://www.biomedcentral.com/1471-2164/14/113 RESEARCH ARTICLE Open Access Systematic analysis of palatal transcriptome to identify cleft palate genes within TGFβ3-knockout mice alleles: RNA-Seq analysis of TGFβ3 Mice Ferhat Ozturk1,4, You Li2, Xiujuan Zhu1, Chittibabu Guda2,3 and Ali Nawshad1* Abstract Background: In humans, cleft palate (CP) accounts for one of the largest number of birth defects with a complex genetic and environmental etiology. TGFβ3 has been established as an important regulator of palatal fusion in mice and it has been shown that TGFβ3-null mice exhibit CP without any other major deformities. However, the genes that regulate cellular decisions and molecular mechanisms maintained by the TGFβ3 pathway throughout palatogenesis are predominantly unexplored. Our objective in this study was to analyze global transcriptome changes within the palate during different gestational ages within TGFβ3 knockout mice to identify TGFβ3-associated genes previously unknown to be associated with the development of cleft palate. We used deep sequencing technology, RNA-Seq, to analyze the transcriptome of TGFβ3 knockout mice at crucial stages of palatogenesis, including palatal growth (E14.5), adhesion (E15.5), and fusion (E16.5). Results: The overall transcriptome analysis of TGFβ3 wildtype mice (C57BL/6) reveals that almost 6000 genes were upregulated during the transition from E14.5 to E15.5 and more than 2000 were downregulated from E15.5 to E16.5.
    [Show full text]
  • Article (Published Version)
    Article The Candidate Schizophrenia Risk Gene DGCR2 Regulates Early Steps of Corticogenesis MOLINARD-CHENU, Aude, DAYER, Alexandre Abstract Alterations in early steps of cortical circuit assembly are thought to play a critical role in vulnerability to schizophrenia (SZ), but the pathogenic impact of SZ-risk mutations on corticogenesis remains to be determined. DiGeorge syndrome critical region 2 (DGCR2) is located in the 22q11.2 locus, whose deletion is a major risk factor for SZ. Moreover, exome sequencing of individuals with idiopathic SZ identified a rare missense mutation in DGCR2, further suggesting that DGCR2 is involved in SZ. Reference MOLINARD-CHENU, Aude, DAYER, Alexandre. The Candidate Schizophrenia Risk Gene DGCR2 Regulates Early Steps of Corticogenesis. Biological Psychiatry, 2018, vol. 83, no. 8, p. 692-706 DOI : 10.1016/j.biopsych.2017.11.015 PMID : 29305086 Available at: http://archive-ouverte.unige.ch/unige:124259 Disclaimer: layout of this document may differ from the published version. 1 / 1 Biological Psychiatry Archival Report The Candidate Schizophrenia Risk Gene DGCR2 Regulates Early Steps of Corticogenesis Aude Molinard-Chenu and Alexandre Dayer ABSTRACT BACKGROUND: Alterations in early steps of cortical circuit assembly are thought to play a critical role in vulnerability to schizophrenia (SZ), but the pathogenic impact of SZ-risk mutations on corticogenesis remains to be determined. DiGeorge syndrome critical region 2 (DGCR2) is located in the 22q11.2 locus, whose deletion is a major risk factor for SZ. Moreover, exome sequencing of individuals with idiopathic SZ identified a rare missense mutation in DGCR2, further suggesting that DGCR2 is involved in SZ. METHODS: Here we investigated the function of Dgcr2 and the pathogenic impact of the SZ-risk DGCR2 mutation in mouse corticogenesis using in utero electroporation targeted to projection neurons.
    [Show full text]
  • Gene Ontology Functional Annotations and Pleiotropy
    Network based analysis of genetic disease associations Sarah Gilman Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy under the Executive Committee of the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014 © 2013 Sarah Gilman All Rights Reserved ABSTRACT Network based analysis of genetic disease associations Sarah Gilman Despite extensive efforts and many promising early findings, genome-wide association studies have explained only a small fraction of the genetic factors contributing to common human diseases. There are many theories about where this “missing heritability” might lie, but increasingly the prevailing view is that common variants, the target of GWAS, are not solely responsible for susceptibility to common diseases and a substantial portion of human disease risk will be found among rare variants. Relatively new, such variants have not been subject to purifying selection, and therefore may be particularly pertinent for neuropsychiatric disorders and other diseases with greatly reduced fecundity. Recently, several researchers have made great progress towards uncovering the genetics behind autism and schizophrenia. By sequencing families, they have found hundreds of de novo variants occurring only in affected individuals, both large structural copy number variants and single nucleotide variants. Despite studying large cohorts there has been little recurrence among the genes implicated suggesting that many hundreds of genes may underlie these complex phenotypes. The question
    [Show full text]
  • Exome Sequencing in Sporadic Autism Spectrum Disorders Identifies Severe De Novo Mutations
    LETTERS Exome sequencing in sporadic autism spectrum disorders identifies severe de novo mutations Brian J O’Roak1, Pelagia Deriziotis2, Choli Lee1, Laura Vives1, Jerrod J Schwartz1, Santhosh Girirajan1, Emre Karakoc1, Alexandra P MacKenzie1, Sarah B Ng1, Carl Baker1, Mark J Rieder1, Deborah A Nickerson1, Raphael Bernier3, Simon E Fisher2,4, Jay Shendure1 & Evan E Eichler1,5 Evidence for the etiology of autism spectrum disorders (ASDs) In contrast with array­based analysis of large de novo copy number has consistently pointed to a strong genetic component variants (CNVs), this approach has greater potential to implicate complicated by substantial locus heterogeneity1,2. We single genes in ASDs. sequenced the exomes of 20 individuals with sporadic ASD We selected 20 trios with an idiopathic ASD, each consistent with a (cases) and their parents, reasoning that these families sporadic ASD based on clinical evaluations (Supplementary Table 1), would be enriched for de novo mutations of major effect. pedigree structure, familial phenotypic evaluation, family history We identified 21 de novo mutations, 11 of which were and/or elevated parental age. Each family was initially screened by protein altering. Protein-altering mutations were significantly array comparative genomic hybridization (CGH) using a customized enriched for changes at highly conserved residues. We microarray9. We identified no large (>250 kb) de novo CNVs but did identified potentially causative de novo events in 4 out of identify a maternally inherited deletion (~350 kb) at 15q11.2 in one 20 probands, particularly among more severely affected family (Supplementary Fig. 1). This deletion has been associated individuals, in FOXP1, GRIN2B, SCN1A and LAMC3.
    [Show full text]
  • Ejhg2009157.Pdf
    European Journal of Human Genetics (2010) 18, 342–347 & 2010 Macmillan Publishers Limited All rights reserved 1018-4813/10 $32.00 www.nature.com/ejhg ARTICLE Fine mapping and association studies of a high-density lipoprotein cholesterol linkage region on chromosome 16 in French-Canadian subjects Zari Dastani1,2,Pa¨ivi Pajukanta3, Michel Marcil1, Nicholas Rudzicz4, Isabelle Ruel1, Swneke D Bailey2, Jenny C Lee3, Mathieu Lemire5,9, Janet Faith5, Jill Platko6,10, John Rioux6,11, Thomas J Hudson2,5,7,9, Daniel Gaudet8, James C Engert*,2,7, Jacques Genest1,2,7 Low levels of high-density lipoprotein cholesterol (HDL-C) are an independent risk factor for cardiovascular disease. To identify novel genetic variants that contribute to HDL-C, we performed genome-wide scans and quantitative association studies in two study samples: a Quebec-wide study consisting of 11 multigenerational families and a study of 61 families from the Saguenay– Lac St-Jean (SLSJ) region of Quebec. The heritability of HDL-C in these study samples was 0.73 and 0.49, respectively. Variance components linkage methods identified a LOD score of 2.61 at 98 cM near the marker D16S515 in Quebec-wide families and an LOD score of 2.96 at 86 cM near the marker D16S2624 in SLSJ families. In the Quebec-wide sample, four families showed segregation over a 25.5-cM (18 Mb) region, which was further reduced to 6.6 Mb with additional markers. The coding regions of all genes within this region were sequenced. A missense variant in CHST6 segregated in four families and, with additional families, we observed a P value of 0.015 for this variant.
    [Show full text]
  • Chicken Fatness: from Qtl to Candidate Gene
    CHICKEN FATNESS: FROM QTL TO CANDIDATE GENE DANYEL JENNEN Promotor: Prof. dr. M.A.M. Groenen Persoonlijk hoogleraar bij de leerstoelgroep Fokkerij en Genetica Wageningen Universiteit Co-promoter: Dr. ing. R.P.M.A. Crooijmans Universitair docent bij de leerstoelgroep Fokkerij en Genetica Wageningen Universiteit Promotiecommissie: Dr. M. Douaire Institut national de la recherche agronomique, Frankrijk Prof. dr. ir. M. Koornneef Wageningen Universiteit Prof. dr. M.R. Müller Wageningen Universiteit Dr. ir. J. Keijer Rikilt, Wageningen Dit onderzoek is uitgevoerd binnen de onderzoekschool WIAS Danyel Gerardus Jacobus Jennen Chicken fatness: From QTL to candidate gene Proefschrift Ter verkrijging van de graad van doctor op gezag van de rector magnificus van Wageningen Universiteit, prof. dr. ir. L. Speelman, in het openbaar te verdedigen op dinsdag 1 juni 2004 des namiddags te vier uur in de Aula D.G.J. Jennen Chicken fatness: From QTL to candidate gene Thesis Wageningen University, The Netherlands, 2004 - with summary in Dutch -176 p ISBN 90-8504-069-8 CONTENTS CHAPTER 1 1 GENERAL INTRODUCTION CHAPTER 2 15 DETECTION AND LOCALIZATION OF QUANTITATIVE TRAIT LOCI AFFECTING FATNESS IN BROILERS CHAPTER 3 35 CONFIRMATION OF QUANTITATIVE TRAIT LOCI AFFECTING FATNESS IN CHICKEN USING AN ADVANCED INTERCROSS LINE CHAPTER 4 57 A COMPARATIVE MAP OF CHICKEN CHROMOSOME 24 AND HUMAN CHROMOSOME 11 CHAPTER 5 73 COMPARATIVE MAP BETWEEN CHICKEN CHROMOSOME 15 AND HUMAN CHROMOSOMAL REGION 12q24 AND 22q11-q12 CHAPTER 6 97 A RADIATION HYBRID MAP OF CHICKEN CHROMOSOME 15 CHAPTER 7 107 IDENTIFICATION AND SNP ANALYSIS OF CANDIDATE GENES FOR FATNESS TRAITS IN CHICKEN CHAPTER 8 125 GENERAL DISCUSSION SUMMARY 145 SAMENVATTING 153 LIST OF PUBLICATIONS 161 DANKWOORD 163 CURRICULUM VITAE 165 TRAINING AND SUPERVISION PLAN WIAS 167 CHAPTER 1 GENERAL INTRODUCTION General Introduction Excessive body fatness has long been of interest to those concerned both with research on human obesity as well as on production in farm animals.
    [Show full text]
  • A Catalog of Hemizygous Variation in 127 22Q11 Deletion Patients
    A catalog of hemizygous variation in 127 22q11 deletion patients. Matthew S Hestand, KU Leuven, Belgium Beata A Nowakowska, KU Leuven, Belgium Elfi Vergaelen, KU Leuven, Belgium Jeroen Van Houdt, KU Leuven, Belgium Luc Dehaspe, UZ Leuven, Belgium Joshua A Suhl, Emory University Jurgen Del-Favero, University of Antwerp Geert Mortier, Antwerp University Hospital Elaine Zackai, The Children's Hospital of Philadelphia Ann Swillen, KU Leuven, Belgium Only first 10 authors above; see publication for full author list. Journal Title: Human Genome Variation Volume: Volume 3 Publisher: Nature Publishing Group: Open Access Journals - Option B | 2016-01-14, Pages 15065-15065 Type of Work: Article | Final Publisher PDF Publisher DOI: 10.1038/hgv.2015.65 Permanent URL: https://pid.emory.edu/ark:/25593/rncxx Final published version: http://dx.doi.org/10.1038/hgv.2015.65 Copyright information: © 2016 Official journal of the Japan Society of Human Genetics This is an Open Access work distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/). Accessed September 28, 2021 7:41 PM EDT OPEN Citation: Human Genome Variation (2016) 3, 15065; doi:10.1038/hgv.2015.65 Official journal of the Japan Society of Human Genetics 2054-345X/16 www.nature.com/hgv ARTICLE A catalog of hemizygous variation in 127 22q11 deletion patients Matthew S Hestand1, Beata A Nowakowska1,2,Elfi Vergaelen1, Jeroen Van Houdt1,3, Luc Dehaspe3, Joshua A Suhl4, Jurgen Del-Favero5, Geert Mortier6, Elaine Zackai7,8, Ann Swillen1, Koenraad Devriendt1, Raquel E Gur8, Donna M McDonald-McGinn7,8, Stephen T Warren4, Beverly S Emanuel7,8 and Joris R Vermeesch1 The 22q11.2 deletion syndrome is the most common microdeletion disorder, with wide phenotypic variability.
    [Show full text]