V12a18-Klintworth Pgmkr

Total Page:16

File Type:pdf, Size:1020Kb

V12a18-Klintworth Pgmkr Molecular Vision 2006; 12:159-76 <http://www.molvis.org/molvis/v12/a18/> ©2006 Molecular Vision Received 13 October 2005 | Accepted 8 March 2006 | Published 10 March 2006 CHST6 mutations in North American subjects with macular corneal dystrophy: a comprehensive molecular genetic review Gordon K. Klintworth,1,2 Clayton F. Smith,2 Brandy L. Bowling2 Departments of 1Pathology and 2Ophthalmology, Duke University Medical Center, Durham, NC Purpose: To evaluate mutations in the carbohydrate sulfotransferase-6 (CHST6) gene in American subjects with macular corneal dystrophy (MCD). Methods: We analyzed CHST6 in 57 patients from 31 families with MCD from the United States, 57 carriers (parents or children), and 27 unaffected blood relatives of affected subjects. We compared the observed nucleotide sequences with those found by numerous investigators in other populations with MCD and in controls. Results: In 24 families, the corneal disorder could be explained by mutations in the coding region of CHST6 or in the region upstream of this gene in both the maternal and paternal chromosome. In most instances of MCD a homozygous or heterozygous missense mutation in exon 3 of CHST6 was found. Six cases resulted from a deletion upstream of CHST6. Conclusions: Nucleotide changes within the coding region of CHST6 are predicted to alter the encoded protein signifi- cantly within evolutionary conserved parts of the encoded sulfotransferase. Our findings support the hypothesis that CHST6 mutations are cardinal to the pathogenesis of MCD. Moreover, the observation that some cases of MCD cannot be explained by mutations in CHST6 suggests that MCD may result from other subtle changes in CHST6 or from genetic heterogeneity. Macular corneal dystrophy (MCD; OMIM 217800), a polysaccharidoses [18], and analyses of sulfated GAGs pro- rare inherited disorder first described in 1890 by Groenouw duced by organ cultures of corneas with MCD led to the dis- [1], has been identified throughout the world. MCD is most covery that corneal tissue with MCD does not synthesize KS prevalent in India [2-4] and Saudi Arabia [5]. In some coun- [19] or normal KS containing proteoglycans (PGs) because of tries, this disease accounts for a high percentage (10-75%) of defective sulfation [20,21]. Immunochemical studies using an the corneal dystrophies requiring keratoplasty [6,7]. Clinically, antibody that recognizes antigenic KS (AgKS) disclosed het- MCD is characterized by a cloudiness of the cornea and ir- erogeneity among cases of MCD based on the reactivity of regularly shaped superficial opacities of both eyes that pro- corneal tissue with the antibody [22]. Subsequently this led to gressively extend through the entire thickness of the central the recognition of three immunophenotypes that are clinically and peripheral corneal stroma. The corneal stroma is thinner and histopathologically indistinguishable from each other than normal [8-10]. In 1938, its autosomal recessive mode of [5,22-24]. Most often neither the serum nor the corneal tissue inheritance became appreciated [11]. More than two decades contain AgKS (MCD type I), but sometimes AgKS is absent later Jones and Zimmerman [12,13] differentiated MCD his- in the corneal stroma and the serum but can be detected in the topathologically from the other two major stromal corneal keratocytes (MCD type IA). A third immunophenotype (MCD dystrophies known as granular and lattice corneal dystrophy. type II) is characterized by the presence of AgKS in corneal Histopathologically, MCD is typified by an intracellular stor- tissue and detectable serum levels of AgKS that are often age of glycosaminoglycans (GAGs) within keratocytes and present in normal amounts. MCD type I corneas have been the corneal endothelium combined with an extracellular depo- found to synthesize an abnormal KS-PG with sition of similar material in the corneal stroma and Descemet’s lactosaminoglycan side chains that lack sulfate. In sharp con- membrane [5,14]. Guttae are common on Descemet’s mem- trast, an MCD type II cornea produced a normal ratio of KS- brane [15]. In 1964, because of similarities to the systemic PG to dermatan sulfate-PG, but the net synthesis of PGs was mucopolysaccharidoses, Klintworth and Vogel [16] suggested below normal [25]. However, the chondroitin/dermatan sul- MCD might be a mucopolysaccharidosis localized to the cor- fate side chains on decorin were sulfated. Subsequently, fluo- nea. A metabolic defect in keratan sulfate (KS), the major cor- rochrome-assisted carbohydrate electrophoresis disclosed that neal GAG [17], was suspected because of the histochemical the KS chain size within the cornea and cartilage in MCD attributes of the corneal accumulations and that the cornea type I was reduced and chain sulfation was absent [26]. In a was the only site of overt abnormalities. Cell culture studies cornea with MCD type II, the sulfation of N-acetylglucosamine disclosed differences from the systemic muco- and galactose was significantly reduced and the chain size was also reduced, but to a lesser degree than in MCD type I [26]. Correspondence to: Gordon K. Klintworth, MD, PhD, Duke Univer- The absence or paucity of sulfate in KS and KS-contain- sity Medical Center, Box 3802, Durham, NC, 27710; Phone: (919) ing PGs in corneas with MCD suggested that patients with 684-3550; FAX: (919) 684-9225; email: [email protected] MCD were deficient in a carbohydrate sulfotransferase that 159 Molecular Vision 2006; 12:159-76 <http://www.molvis.org/molvis/v12/a18/> ©2006 Molecular Vision catalyzes the transfer of sulfate groups from 3'- Because the gene responsible for MCD was suspected of phosphoadenosine 5'-phosphosulfate (PAPS) to KS in a man- encoding for a carbohydrate sulfotransferase and had been fine ner comparable to which sulfate esters are put on other en- mapped to the same region as CHST5 and CHST6, these genes dogenous and exogenous substrates (PAPS + R-OH ->PAP + became logical suspects for the MCD disease gene. In 2000, R-OSO3). An analysis of the serum in patients with MCD type Akama et al. [33] discovered mutations in CHST6 in MCD I disclosed normal levels of enzymatic activity for sulfating at and also found insertional or deletional defects in the region least one of the two sugars present in KS and an enzyme defi- between CHST5 and CHST6 in some cases. Subsequently, cient for sulfating N-acetylglucosamine (GlcNAc) was thought several laboratories confirmed these observations [2-4,39-52]. to be present [27]. Because the serum KS is considered to be This report documents an analysis of CHST6 in 57 pa- derived from cartilage, the discovery of undetectable levels of tients from 31 families from the United States with MCD, and AgKS in the serum of individuals with MCD type I suggested reviews these findings with all previously documented muta- that the KS sulfotransferase deficiency was not restricted to tions. the cornea. Direct evidence of cartilage involvement was ob- tained later in cartilage from the nose and ear in MCD type I METHODS [26,28]. Cartilage lacks abnormal accumulations comparable Subjects: We analyzed CHST6 in 141 individuals (57 affected to those in the cornea, but the chondrocytes and extracellular patients, 57 carriers [parents or children], and 27 normal blood matrix do not contain AgKS and the KS content of cartilage is related family members) from 31 families with MCD. In all at least 800 times lower than normal [28]. instances, the diagnosis of MCD was made on a combination Using families from Iceland, where MCD was the most of the characteristic clinical features together with the typical frequent indication for penetrating keratoplasty [7], the gene histopathologic findings in the corneal tissue obtained follow- for MCD type I was mapped to chromosome 16 (16q22) and ing a penetrating keratoplasty in one or both eyes. All patients linkage data hinted that MCD type II was also at this locus were born in the United states and some ancestral lines could [29]. Fine mapping refined the location of the MCD gene. be traced to England, Ireland, Germany, Holland, Northern Two candidate genes (TAT and LCAT) in that part of chromo- Ireland, Scotland, Switzerland, and Norway. Nonmolecular some 16 were excluded [30-32]. genetic studies have been previously reported on some of these A sulfotransferase suspected of being defective in MCD cases [14,16,18,19,53-56]. This study was approved by the would belong to the galactose/N-acetylgalactosamine/N- Internal Review Board of Duke University and conformed to acetylglucosamine 6-O-sulfotransferase family of carbohy- the tenets of the declaration of Helsinki. Written informed drate sulfotransferases. This family of enzymes catalyzes 6- consent was obtained from all participants. To evaluate the O-sulfation on the 6-hydroxyl of GlcNAc, galactose (GAL), CHST6 gene in 11 deceased or unavailable patients from five or N-acetylgalactosamine (GalNAc) [33]. All known families with MCD, we analyzed the DNA of surviving sib- sulfotransferases contain highly conserved regions [34], in- lings, parents, and children to establish which mutations had cluding a 5'-PSB (phosphosulfate-binding) and a 3'-PB (3'- been transmitted to the carriers. We also analyzed DNA from phosphate binding domain) [35]. The carbohydrate 50 control specimens from the United States and DNA from sulfotransferases in different species possess marked sequence 10 normal spouses who married into these families. similarities at the amino acid level particularly in their cata- Determination of MCD immunophenotypes: Prior to mo- lytic domains and, with the exception of CHST3 (also known lecular genetic studies, the immunophenotypes had been de- as GST-0), the coding sequences of the open reading frames termined in 32 of the 57 subjects with MCD by ascertaining (ORFs) for all CHST genes are within a single exon [36]. The the presence or absence of sulfated epitopes in KS in the cor- galactose/N-acetylgalactosamine/N-acetylglucosamine 6-O- neal tissue and/or serum.
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]
  • 1 Mutational Heterogeneity in Cancer Akash Kumar a Dissertation
    Mutational Heterogeneity in Cancer Akash Kumar A dissertation Submitted in partial fulfillment of requirements for the degree of Doctor of Philosophy University of Washington 2014 June 5 Reading Committee: Jay Shendure Pete Nelson Mary Claire King Program Authorized to Offer Degree: Genome Sciences 1 University of Washington ABSTRACT Mutational Heterogeneity in Cancer Akash Kumar Chair of the Supervisory Committee: Associate Professor Jay Shendure Department of Genome Sciences Somatic mutation plays a key role in the formation and progression of cancer. Differences in mutation patterns likely explain much of the heterogeneity seen in prognosis and treatment response among patients. Recent advances in massively parallel sequencing have greatly expanded our capability to investigate somatic mutation. Genomic profiling of tumor biopsies could guide the administration of targeted therapeutics on the basis of the tumor’s collection of mutations. Central to the success of this approach is the general applicability of targeted therapies to a patient’s entire tumor burden. This requires a better understanding of the genomic heterogeneity present both within individual tumors (intratumoral) and amongst tumors from the same patient (intrapatient). My dissertation is broadly organized around investigating mutational heterogeneity in cancer. Three projects are discussed in detail: analysis of (1) interpatient and (2) intrapatient heterogeneity in men with disseminated prostate cancer, and (3) investigation of regional intratumoral heterogeneity in
    [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]
  • 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]
  • Genomic and Transcriptome Analysis Revealing an Oncogenic Functional Module in Meningiomas
    Neurosurg Focus 35 (6):E3, 2013 ©AANS, 2013 Genomic and transcriptome analysis revealing an oncogenic functional module in meningiomas XIAO CHANG, PH.D.,1 LINGLING SHI, PH.D.,2 FAN GAO, PH.D.,1 JONATHAN RUssIN, M.D.,3 LIYUN ZENG, PH.D.,1 SHUHAN HE, B.S.,3 THOMAS C. CHEN, M.D.,3 STEVEN L. GIANNOTTA, M.D.,3 DANIEL J. WEISENBERGER, PH.D.,4 GAbrIEL ZADA, M.D.,3 KAI WANG, PH.D.,1,5,6 AND WIllIAM J. MAck, M.D.1,3 1Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California; 2GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China; 3Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California; 4USC Epigenome Center, Keck School of Medicine, University of Southern California, Los Angeles, California; 5Department of Psychiatry, Keck School of Medicine, University of Southern California, Los Angeles, California; and 6Division of Bioinformatics, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California Object. Meningiomas are among the most common primary adult brain tumors. Although typically benign, roughly 2%–5% display malignant pathological features. The key molecular pathways involved in malignant trans- formation remain to be determined. Methods. Illumina expression microarrays were used to assess gene expression levels, and Illumina single- nucleotide polymorphism arrays were used to identify copy number variants in benign, atypical, and malignant me- ningiomas (19 tumors, including 4 malignant ones). The authors also reanalyzed 2 expression data sets generated on Affymetrix microarrays (n = 68, including 6 malignant ones; n = 56, including 3 malignant ones).
    [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]
  • Construction of a Natural Panel of 11P11.2 Deletions and Further Delineation of the Critical Region Involved in Potocki–Shaffer Syndrome
    European Journal of Human Genetics (2005) 13, 528–540 & 2005 Nature Publishing Group All rights reserved 1018-4813/05 $30.00 www.nature.com/ejhg ARTICLE Construction of a natural panel of 11p11.2 deletions and further delineation of the critical region involved in Potocki–Shaffer syndrome Keiko Wakui1,12, Giuliana Gregato2,3,12, Blake C Ballif2, Caron D Glotzbach2,4, Kristen A Bailey2,4, Pao-Lin Kuo5, Whui-Chen Sue6, Leslie J Sheffield7, Mira Irons8, Enrique G Gomez9, Jacqueline T Hecht10, Lorraine Potocki1,11 and Lisa G Shaffer*,2,4 1Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA; 2Health Research and Education Center, Washington State University, Spokane, WA, USA; 3Dip. Patologia Umana ed Ereditaria, Sez. Biologia Generale e Genetica Medica, Universita` degli Studi di Pavia, Italy; 4Sacred Heart Medical Center, Spokane, WA, USA; 5Department of Obstetrics and Gynecology, National Cheng-Kung University Medical College, Taiwan; 6Department of Pediatrics, Taipei Municipal Women and Children’s Hospital, Taiwan; 7Genetic Health Services Victoria, Murdoch Children’s Research Institute, Department of Paediatrics, University of Melbourne, Victoria, Australia; 8Division of Genetics, Department of Medicine, Children’s Hospital, Harvard Medical School, Boston, MA, USA; 9Area de Gene´tica, Centro de Desarrollo Infantil y Departamento de Pediatrı´a Hospital Materno Infantil-Hospital Regional Universitario ‘Infanta Cristina’, Badajoz, Spain; 10Department of Pediatrics, University of Texas Medical School at Houston, TX, USA; 11Texas Children’s Hospital, Houston, TX, USA Potocki–Shaffer syndrome (PSS) is a contiguous gene deletion syndrome that results from haploinsufficiency of at least two genes within the short arm of chromosome 11[del(11)(p11.2p12)].
    [Show full text]
  • Downloaded from Here
    bioRxiv preprint doi: https://doi.org/10.1101/017566; this version posted November 19, 2015. 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-NC-ND 4.0 International license. 1 1 Testing for ancient selection using cross-population allele 2 frequency differentiation 1;∗ 3 Fernando Racimo 4 1 Department of Integrative Biology, University of California, Berkeley, CA, USA 5 ∗ E-mail: [email protected] 6 1 Abstract 7 A powerful way to detect selection in a population is by modeling local allele frequency changes in a 8 particular region of the genome under scenarios of selection and neutrality, and finding which model is 9 most compatible with the data. Chen et al. [2010] developed a composite likelihood method called XP- 10 CLR that uses an outgroup population to detect departures from neutrality which could be compatible 11 with hard or soft sweeps, at linked sites near a beneficial allele. However, this method is most sensitive 12 to recent selection and may miss selective events that happened a long time ago. To overcome this, 13 we developed an extension of XP-CLR that jointly models the behavior of a selected allele in a three- 14 population tree. Our method - called 3P-CLR - outperforms XP-CLR when testing for selection that 15 occurred before two populations split from each other, and can distinguish between those events and 16 events that occurred specifically in each of the populations after the split.
    [Show full text]
  • A Comprehensive Evaluation of 181 Reported CHST6 Variants in Patients with Macular Corneal Dystrophy
    www.aging‐us.com AGING 2019, Vol. 11, No. 3 Research Paper A comprehensive evaluation of 181 reported CHST6 variants in patients with macular corneal dystrophy Jing Zhang1, Dan Wu1, Yue Li1, Yidan Fan1, Yiqin Dai1, Jianjiang Xu1 1Department of Ophthalmology and Visual Science, Eye and ENT Hospital, Shanghai Medical College of Fudan University, NHC Key Laboratory of Myopia (Fudan University), Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China Correspondence to: Jianjiang Xu; email: [email protected] Keywords: CHST6, macular corneal dystrophy, genetic variants Received: October 23, 2018 Accepted: January 25, 2019 Published: February 4, 2019 Copyright: Zhang et al. This is an open‐access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Macular corneal dystrophy (MCD) is an autosomal recessive disease featured by bilateral progressive stromal clouding and loss of vision, consequently necessitating corneal transplantation. Variants in CHST6 gene have been recognized as the most critical genetic components in MCD. Although many CHST6 variants have been described until now, the detailed mechanisms underlying MCD are still far from understood. In this study, we integrated all the reported CHST6 variants described in 408 MCD cases, and performed a comprehensive evaluation to better illustrate the causality of these variants. The results showed that majority of these variants (165 out of 181) could be classified as pathogenic or likely pathogenic. Interestingly, we also identified several disease causal variants with ethnic specificity. In addition, the results underscored the strong correlation between mutant frequency and residue conservation in the general population (Spearman’s correlation coefficient = ‐0.311, P = 1.20E‐05), thus providing potential candidate targets for further genetic manipulation.
    [Show full text]
  • Dynamic Changes in the Epigenomic Landscape Regulate Human Organogenesis and Link to Developmental Disorders
    ARTICLE https://doi.org/10.1038/s41467-020-17305-2 OPEN Dynamic changes in the epigenomic landscape regulate human organogenesis and link to developmental disorders Dave T. Gerrard 1,6, Andrew A. Berry1,6, Rachel E. Jennings1,2, Matthew J. Birket 1, Peyman Zarrineh1, Myles G. Garstang1, Sarah L. Withey1, Patrick Short 3, Sandra Jiménez-Gancedo4, Panos N. Firbas4, Ian Donaldson 1, Andrew D. Sharrocks1, Karen Piper Hanley 1,5, Matthew E. Hurles3, ✉ José Luis Gomez-Skarmeta4, Nicoletta Bobola1 & Neil A. Hanley 1,2 1234567890():,; How the genome activates or silences transcriptional programmes governs organ formation. Little is known in human embryos undermining our ability to benchmark the fidelity of stem cell differentiation or cell programming, or interpret the pathogenicity of noncoding variation. Here, we study histone modifications across thirteen tissues during human organogenesis. We integrate the data with transcription to build an overview of how the human genome differentially regulates alternative organ fates including by repression. Promoters from nearly 20,000 genes partition into discrete states. Key developmental gene sets are actively repressed outside of the appropriate organ without obvious bivalency. Candidate enhancers, functional in zebrafish, allow imputation of tissue-specific and shared patterns of transcrip- tion factor binding. Overlaying more than 700 noncoding mutations from patients with developmental disorders allows correlation to unanticipated target genes. Taken together, the data provide a comprehensive genomic framework for investigating normal and abnormal human development. 1 Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK. 2 Endocrinology Department, Manchester University NHS Foundation Trust, Grafton Street, Manchester M13 9WU, UK.
    [Show full text]
  • Insertion Variants Missing in the Human Reference Genome Are Widespread Among Human Populations Young-Gun Lee1†, Jin-Young Lee2†, Junhyong Kim3 and Young-Joon Kim1,2*
    Lee et al. BMC Biology (2020) 18:167 https://doi.org/10.1186/s12915-020-00894-1 RESEARCH ARTICLE Open Access Insertion variants missing in the human reference genome are widespread among human populations Young-gun Lee1†, Jin-young Lee2†, Junhyong Kim3 and Young-Joon Kim1,2* Abstract Background: Structural variants comprise diverse genomic arrangements including deletions, insertions, inversions, and translocations, which can generally be detected in humans through sequence comparison to the reference genome. Among structural variants, insertions are the least frequently identified variants, mainly due to ascertainment bias in the reference genome, lack of previous sequence knowledge, and low complexity of typical insertion sequences. Though recent developments in long-read sequencing deliver promise in annotating individual non-reference insertions, population-level catalogues on non-reference insertion variants have not been identified and the possible functional roles of these hidden variants remain elusive. Results: To detect non-reference insertion variants, we developed a pipeline, InserTag, which generates non- reference contigs by local de novo assembly and then infers the full-sequence of insertion variants by tracing contigs from non-human primates and other human genome assemblies. Application of the pipeline to data from 2535 individuals of the 1000 Genomes Project helped identify 1696 non-reference insertion variants and re-classify the variants as retention of ancestral sequences or novel sequence insertions based on the ancestral state. Genotyping of the variants showed that individuals had, on average, 0.92-Mbp sequences missing from the reference genome, 92% of the variants were common (allele frequency > 5%) among human populations, and more than half of the variants were major alleles.
    [Show full text]