Investigating the Genetic Basis of Cisplatin-Induced Ototoxicity in Adult South African Patients

Total Page:16

File Type:pdf, Size:1020Kb

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 ………………………………………. 14 1.3.2.1 Cisplatin-induced ototoxicity ………………………………………………... 14 1.3.2.2 Mechanism of cisplatin-induced ototoxicity ………………………………. 16 1.4 Audiometry ………………………………………………………………………. 19 1.5 Risk factors of ototoxicity ………………………………………………………. 20 1.6 Pharmacogenetics ………………………………………………………………. 21 1.6.1 The pharmacogenetics of cisplatin-induced ototoxicity ……………………. 21 1.6.1.1 Genes involved in drug metabolism and detoxification ………………….. 23 1.6.1.1.1 The glutathione S-transferases …………………………………………. 23 1.6.1.1.2 Methyltransferases ……………………………………………………….. 24 1.6.1.1.3 Dehydrogenases …………………………………………………………... 25 1.6.1.2 Membrane receptors and transporters ……………………………………. 25 1.6.1.3 Deafness-associated genes ……………………………………………….. 26 1.6.1.4 Other genes ………………………………………………………………….. 27 1.7 Future considerations or directions …………………………………………….. 28 1.8 Research aim and objectives …………………………………………………… 29 2. Analysis of clinical and demographic risk factors of cisplatin-induced ototoxicity …... 30 2.1 Introduction ……………………………………………………………………….. 30 2.2 Methods …………………………………………………………………………… 31 2.2.1 Patients …………………………………………………………………………. 31 2.2.2 Treatment details ………………………………………………………………. 32 2.2.3 Audiometric monitoring ………………………………………………………... 32 2.2.4 Statistical analyses …………………………………………………………….. 33 2.3 Results …………………………………………………………………………….. 33 3 2.3.1 Patient cohort …………………………………………………………………... 33 2.3.2 Association of clinical characteristics with ototoxicity ……………………… 34 2.3.3 Association of demographic variables with ototoxicity …………………….. 35 2.3.4 Statistical modelling of cisplatin-induced ototoxicity ……………………….. 37 2.4 Discussion ………………………………………………………………………… 38 2.5 Conclusion ………………………………………………………………………… 40 3. Investigation of candidate variants and their role in cisplatin-induced ototoxicity …… 42 3.1 Introduction ……………………………………………………………………….. 42 3.2 Methods …………………………………………………………………………… 45 3.2.1 Research design ……………………………………………………………….. 45 3.2.2 Patient recruitment …………………………………………………………….. 45 3.2.2.1 Inclusion and exclusion criteria …………………………………………….. 45 3.2.2.2 Collection of data, biological samples, and informed consent ………….. 46 3.2.2.3 Audiometric analysis ………………………………………………………… 46 3.2.3 DNA extraction and quality control ………………………………………….. 47 3.2.4 Candidate selection and the amplification of target DNA regions ………... 47 3.2.4.1 PCR conditions ………………………………………………………………. 48 3.2.4.2 Purification of PCR products ………………………………………………. 49 3.2.5 SNaPshot® genotyping ……………………………………………………….. 49 3.2.5.1 Visualisation and analysis of SNaPshot® reaction products ……………. 50 3.2.6 Validation of genotyping ………………………………………………………. 50 3.2.6.1 Direct cycle sequencing reaction conditions ……………………………… 51 3.2.6.2 Analysis of sequencing products …………………………………………... 51 3.2.7 Statistical analysis of data …………………………………………………….. 52 3.3 Results …………………………………………………………………………….. 53 3.3.1 Patient cohort …………………………………………………………………... 53 3.3.2 Incidence of ototoxicity in the cohort ………………………………………… 54 3.3.3 Candidate gene study …………………………………………………………. 56 3.3.3.1 Single site analysis ………………………………………………………….. 56 3.3.3.2 Haplotype analysis …………………………………………………………... 61 3.3.3.3 Regression modelling of clinical, demographic and genetic variables ……………………………………………………………………………….. 62 3.3.3.4 Other markers of treatment response ……………………………………... 64 3.4 Discussion ………………………………………………………………………… 65 3.5 Conclusion ………………………………………………………………………… 70 4. Whole-exome sequencing to identify potential modifiers of drug response …………. 71 4.1 Introduction ……………………………………………………………………….. 71 4 4.2 Methods …………………………………………………………………………… 73 4.2.1 Patients and samples ………………………………………………………… 73 4.2.2 WES ……………………………………………………………………………. 73 4.2.3 Variant annotation ……………………………………………………………... 74 4.2.4 Scoring and filtering of exonic variants ……………………………………… 75 4.2.4.1 Variant-level analysis ……………………………………………………….. 75 4.2.4.1.1 Analysis of gene-disease relationships …………………………………. 75 4.2.4.1.2 Gene prioritisation …………………………………………………………. 76 4.2.4.2 Gene-level analysis …………………………………………………………. 76 4.2.4.2.1 Pathway analysis …………………………………………………………. 76 4.2.4.2.2 Panel analysis ……………………………………………………………... 77 4.2.5 Statistical analysis of patient demographic and genomic data …………… 77 4.3 Results …………………………………………………………………………….. 78 4.3.1 Extreme-phenotype cohort ……………………………………………………. 78 4.3.2 WES ……………………………………………………………………………... 83 4.3.2.1 Principal component analysis ………………………………………………. 86 4.3.2.2 Filtering of variants …………………………………………………………. 88 4.3.2.2.1 Variant-based filtering …………………………………………………….. 91 4.3.2.2.2 Gene-based filtering ………………………………………………………. 92 4.3.2.2.2.1 Panel-based gene-level analysis ……………………………………… 96 4.3.2.2.2.2 Pathway gene-level analysis ………………………………………….. 98 4.4 Discussion ………………………………………………………………………… 101 4.5 Conclusion ………………………………………………………………………… 111 5. Conclusion and future perspectives ……………………………………………………… 112 6. References ………………………………………………………………………………….. 114 6.1 Literature cited …………………………………………………………………… 114 6.2 Websites accessed ………………………………………………………………. 133 Appendices …………………………………………………………………………………….. 136 Appendix A: Letter of ethical approval …………………………………………….. 136 Appendix B: Molecular study information sheet provided to patients during recruitment …………………………………………………………………….. 137 Appendix C: Informed consent form for access to patient records ……………… 139 Appendix D: Informed consent for the collection, analysis and storage of patient biological samples ………………………………………………. 140 Appendix E: Letter of GSH approval ……………………………………………….. 143 Appendix F: Analysis of threshold changes in all prospective and retrospective patients ………………………………………………………………… 144 5 Appendix G: DNA extraction protocols …………………………………………….. 146 Appendix H: Buffers and reagents ………………………………………………….. 148 Appendix I: Tests of association and regression between genotypic and allelic information and ototoxicity in the total patient cohort ………………… 150 Appendix J: Gene panels used in analysis of WES data ………………………… 154 Appendix K: Gene-disease relationships for WES data ………………………….. 165 Appendix L: Panel-based analysis of all unique variants scoring ≥ 4 …………... 169 Appendix M: Results of pathway analysis …………………………………………. 174 Appendix N: Pathway panels ……………………………………………………….. 176 Acknowledgements …………………………………………………………………………… 193 Turnitin: cover page …………………………………………………………………………… 194 Turnitin: summary of originality report ………………………………………………………. 195 6 Abbreviations 5-FU: 5-fluorouracil A: adenine ABC: ATP-binding cassette ACYP2: acylphosphatase 2, muscle type ADA: adenosine deaminase ADME: drug absorption, distribution, metabolism and elimination ADR: adverse drug reaction AFR: 1000 Genomes Project African population AMPK: AMP-activated protein kinase AMR: 1000 Genomes Project admixed American population AP: alkaline phosphatase AQP: aquaporin ARE: antioxidant response element Arntl: aryl hydrocarbon receptor nuclear translocator-like ASHA: American Speech-Language-Hearing Association ASN: 1000 Genomes Project East Asian population ATP: adenosine triphosphate BA: black African BCL2: B-cell CLL/lymphoma 2 bp: base pairs C: cytosine C2: complement component 2 C3: complement component 3 C5: complement component 5 CAF: Central Analytical Facility cAMP: cyclic adenosine monophosphate CAT: catalase Cau: Caucasian CCND1: cyclin D1 CDH13: cadherin 13 CFB: complement factor B CFTR: cystic fibrosis transmembrane conductance regulator CI: confidence interval 1 COCH: cochlin COMT: catechol-O-methyltransferase
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]
  • 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 Identities in Evs Isolated from U87-MG GBM Cells As Determined by NG LC-MS/MS
    Protein identities in EVs isolated from U87-MG GBM cells as determined by NG LC-MS/MS. No. Accession Description Σ Coverage Σ# Proteins Σ# Unique Peptides Σ# Peptides Σ# PSMs # AAs MW [kDa] calc. pI 1 A8MS94 Putative golgin subfamily A member 2-like protein 5 OS=Homo sapiens PE=5 SV=2 - [GG2L5_HUMAN] 100 1 1 7 88 110 12,03704523 5,681152344 2 P60660 Myosin light polypeptide 6 OS=Homo sapiens GN=MYL6 PE=1 SV=2 - [MYL6_HUMAN] 100 3 5 17 173 151 16,91913397 4,652832031 3 Q6ZYL4 General transcription factor IIH subunit 5 OS=Homo sapiens GN=GTF2H5 PE=1 SV=1 - [TF2H5_HUMAN] 98,59 1 1 4 13 71 8,048185945 4,652832031 4 P60709 Actin, cytoplasmic 1 OS=Homo sapiens GN=ACTB PE=1 SV=1 - [ACTB_HUMAN] 97,6 5 5 35 917 375 41,70973209 5,478027344 5 P13489 Ribonuclease inhibitor OS=Homo sapiens GN=RNH1 PE=1 SV=2 - [RINI_HUMAN] 96,75 1 12 37 173 461 49,94108966 4,817871094 6 P09382 Galectin-1 OS=Homo sapiens GN=LGALS1 PE=1 SV=2 - [LEG1_HUMAN] 96,3 1 7 14 283 135 14,70620005 5,503417969 7 P60174 Triosephosphate isomerase OS=Homo sapiens GN=TPI1 PE=1 SV=3 - [TPIS_HUMAN] 95,1 3 16 25 375 286 30,77169764 5,922363281 8 P04406 Glyceraldehyde-3-phosphate dehydrogenase OS=Homo sapiens GN=GAPDH PE=1 SV=3 - [G3P_HUMAN] 94,63 2 13 31 509 335 36,03039959 8,455566406 9 Q15185 Prostaglandin E synthase 3 OS=Homo sapiens GN=PTGES3 PE=1 SV=1 - [TEBP_HUMAN] 93,13 1 5 12 74 160 18,68541938 4,538574219 10 P09417 Dihydropteridine reductase OS=Homo sapiens GN=QDPR PE=1 SV=2 - [DHPR_HUMAN] 93,03 1 1 17 69 244 25,77302971 7,371582031 11 P01911 HLA class II histocompatibility antigen,
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Investigation of Candidate Genes and Mechanisms Underlying Obesity
    Prashanth et al. BMC Endocrine Disorders (2021) 21:80 https://doi.org/10.1186/s12902-021-00718-5 RESEARCH ARTICLE Open Access Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules G. Prashanth1 , Basavaraj Vastrad2 , Anandkumar Tengli3 , Chanabasayya Vastrad4* and Iranna Kotturshetti5 Abstract Background: Obesity associated type 2 diabetes mellitus is a metabolic disorder ; however, the etiology of obesity associated type 2 diabetes mellitus remains largely unknown. There is an urgent need to further broaden the understanding of the molecular mechanism associated in obesity associated type 2 diabetes mellitus. Methods: To screen the differentially expressed genes (DEGs) that might play essential roles in obesity associated type 2 diabetes mellitus, the publicly available expression profiling by high throughput sequencing data (GSE143319) was downloaded and screened for DEGs. Then, Gene Ontology (GO) and REACTOME pathway enrichment analysis were performed. The protein - protein interaction network, miRNA - target genes regulatory network and TF-target gene regulatory network were constructed and analyzed for identification of hub and target genes. The hub genes were validated by receiver operating characteristic (ROC) curve analysis and RT- PCR analysis. Finally, a molecular docking study was performed on over expressed proteins to predict the target small drug molecules. Results: A total of 820 DEGs were identified between
    [Show full text]
  • Effects of Neonatal Stress and Morphine on Murine Hippocampal Gene Expression
    0031-3998/11/6904-0285 Vol. 69, No. 4, 2011 PEDIATRIC RESEARCH Printed in U.S.A. Copyright © 2011 International Pediatric Research Foundation, Inc. Effects of Neonatal Stress and Morphine on Murine Hippocampal Gene Expression SANDRA E. JUUL, RICHARD P. BEYER, THEO K. BAMMLER, FEDERICO M. FARIN, AND CHRISTINE A. GLEASON Department of Pediatrics [S.E.J., C.A.G.], Department of Environmental and Occupational Health Sciences [R.P.B., T.K.B., F.M.F.], University of Washington, Seattle, Washington 98195 ABSTRACT: Critically ill preterm infants experience multiple to severe impairment occurring in close to 50% of ex- stressors while hospitalized. Morphine is commonly prescribed to tremely LBW infants (4). Autism, attention deficit disorder, ameliorate their pain and stress. We hypothesized that neonatal and school failure also occur more frequently in NICU stress will have a dose-dependent effect on hippocampal gene survivors (5). Although some degree of impairment might expression, and these effects will be altered by morphine treat- ment. Male C57BL/6 mice were exposed to five treatment condi- be inevitable, it is likely that the stress and treatments these tions between postnatal d 5 and 9: 1) control, 2) mild stress ϩ infants undergo impact neurologic outcome. Improved un- saline, 3) mild stress ϩ morphine, 4) severe stress ϩ saline, and derstanding of these factors will provide the basis of better 5) severe stress ϩ morphine. Hippocampal RNA was extracted treatments and subsequent improvement in outcomes. and analyzed using Affymetrix Mouse Gene 1.0 ST Arrays. Single Many preterm infants receive opiates for sedation or gene analysis and gene set analysis were used to compare groups analgesia during their NICU stay.
    [Show full text]
  • Enzyme DHRS7
    Toward the identification of a function of the “orphan” enzyme DHRS7 Inauguraldissertation zur Erlangung der Würde eines Doktors der Philosophie vorgelegt der Philosophisch-Naturwissenschaftlichen Fakultät der Universität Basel von Selene Araya, aus Lugano, Tessin Basel, 2018 Originaldokument gespeichert auf dem Dokumentenserver der Universität Basel edoc.unibas.ch Genehmigt von der Philosophisch-Naturwissenschaftlichen Fakultät auf Antrag von Prof. Dr. Alex Odermatt (Fakultätsverantwortlicher) und Prof. Dr. Michael Arand (Korreferent) Basel, den 26.6.2018 ________________________ Dekan Prof. Dr. Martin Spiess I. List of Abbreviations 3α/βAdiol 3α/β-Androstanediol (5α-Androstane-3α/β,17β-diol) 3α/βHSD 3α/β-hydroxysteroid dehydrogenase 17β-HSD 17β-Hydroxysteroid Dehydrogenase 17αOHProg 17α-Hydroxyprogesterone 20α/βOHProg 20α/β-Hydroxyprogesterone 17α,20α/βdiOHProg 20α/βdihydroxyprogesterone ADT Androgen deprivation therapy ANOVA Analysis of variance AR Androgen Receptor AKR Aldo-Keto Reductase ATCC American Type Culture Collection CAM Cell Adhesion Molecule CYP Cytochrome P450 CBR1 Carbonyl reductase 1 CRPC Castration resistant prostate cancer Ct-value Cycle threshold-value DHRS7 (B/C) Dehydrogenase/Reductase Short Chain Dehydrogenase Family Member 7 (B/C) DHEA Dehydroepiandrosterone DHP Dehydroprogesterone DHT 5α-Dihydrotestosterone DMEM Dulbecco's Modified Eagle's Medium DMSO Dimethyl Sulfoxide DTT Dithiothreitol E1 Estrone E2 Estradiol ECM Extracellular Membrane EDTA Ethylenediaminetetraacetic acid EMT Epithelial-mesenchymal transition ER Endoplasmic Reticulum ERα/β Estrogen Receptor α/β FBS Fetal Bovine Serum 3 FDR False discovery rate FGF Fibroblast growth factor HEPES 4-(2-Hydroxyethyl)-1-Piperazineethanesulfonic Acid HMDB Human Metabolome Database HPLC High Performance Liquid Chromatography HSD Hydroxysteroid Dehydrogenase IC50 Half-Maximal Inhibitory Concentration LNCaP Lymph node carcinoma of the prostate mRNA Messenger Ribonucleic Acid n.d.
    [Show full text]
  • A 1Bp Deletion in the Dual Reading Frame Deafness Gene LRTOMT Causes a Frameshift from the First Into the Second Reading Frame
    RESEARCH LETTER A 1 bp Deletion in the Dual Reading Frame Deafness Gene LRTOMT Causes a Frameshift From the First Into the Second Reading Frame Maarten Vanwesemael,1 Isabelle Schrauwen,1 Ruben Ceuppens,1 Fatemeh Alasti,1 Ellen Jorssen,1 Effat Farrokhi,2 Morteza Hashemzadeh Chaleshtori,2 and Guy Van Camp1* 1Department of Medical Genetics, University of Antwerp, 2610, Wilrijk, Belgium 2Cellular and Molecular Research Center, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran Received 29 November 2010; Accepted 12 April 2011 TO THE EDITOR: How to Cite this Article: Congenital hearing impairment (HI) affects one in 650 newborns Vanwesemael M, Schrauwen I, Ceuppens R, and has a genetic cause in 50% of the cases. Autosomal recessive Alasti F, Jorssen E, Farrokhi E, Chaleshtori non-syndromic hearing impairment (ARNSHI) is responsible for MH, Van Camp G. 2011. A 1 bp deletion in the 70–80% of all hereditary cases of HI. ARNSHI is genetically highly dual reading frame deafness gene LRTOMT heterogeneous and until today mutations in 37 genes have been causes a frameshift from the first into the identified [Van Camp and Smith, 2011]. One of the latest genes for second reading frame. ARNSHI is LRTOMT (at locus DFNB63 on 11q13.3-q13.4). The Am J Med Genet Part A 155:2021–2023. structure of human LRTOMT has only recently been elucidated [Ahmed et al., 2008]. LRTOMT is an evolutionary recent fusion gene with alternative reading frames that exclusively exists in analysis was performed on an ABI 3130XL DNA sequence primates and is a fusion of non-primate Lrrc51 (Leucine rich repeat analyzer (Applied Biosystems Inc., Foster City, CA), using standard containing 51) and Tomt (Transmembrane O-methyltransferase) procedures.
    [Show full text]
  • Anti-VPS4A Antibody (ARG43067)
    Product datasheet [email protected] ARG43067 Package: 50 μg anti-VPS4A antibody Store at: -20°C Summary Product Description Rabbit Polyclonal antibody recognizes VPS4A Tested Reactivity Hu, Ms, Rat Tested Application FACS, IHC-P, WB Host Rabbit Clonality Polyclonal Isotype IgG Target Name VPS4A Antigen Species Human Immunogen Recombinant protein corresponding to M1-K71 of Human VPS4A. Conjugation Un-conjugated Alternate Names SKD2; SKD1; hVPS4; Protein SKD2; SKD1A; Vacuolar protein sorting-associated protein 4A; VPS4-1; EC 3.6.4.6; VPS4 Application Instructions Application table Application Dilution FACS 1:150 - 1:500 IHC-P 1:200 - 1:1000 WB 1:500 - 1:2000 Application Note IHC-P: Antigen Retrieval: Heat mediation was performed in EDTA buffer (pH 8.0). * The dilutions indicate recommended starting dilutions and the optimal dilutions or concentrations should be determined by the scientist. Calculated Mw 49 kDa Properties Form Liquid Purification Affinity purification with immunogen. Buffer 0.2% Na2HPO4, 0.9% NaCl, 0.05% Sodium azide and 4% Trehalose. Preservative 0.05% Sodium azide Stabilizer 4% Trehalose Concentration 0.5 mg/ml Storage instruction For continuous use, store undiluted antibody at 2-8°C for up to a week. For long-term storage, aliquot and store at -20°C or below. Storage in frost free freezers is not recommended. Avoid repeated freeze/thaw cycles. Suggest spin the vial prior to opening. The antibody solution should be gently mixed www.arigobio.com 1/3 before use. Note For laboratory research only, not for drug, diagnostic or other use. Bioinformation Gene Symbol VPS4A Gene Full Name vacuolar protein sorting 4 homolog A (S.
    [Show full text]
  • ZFYVE19 (E-14): Sc-165940
    SAN TA C RUZ BI OTEC HNOL OG Y, INC . ZFYVE19 (E-14): sc-165940 BACKGROUND APPLICATIONS Zinc-finger proteins contain DNA-binding domains and have a wide variety of ZFYVE19 (E-14) is recommended for detection of ZFYVE19 of mouse, rat and functions, most of which encompass some form of transcriptional activation human origin by Western Blotting (starting dilution 1:200, dilution range or repression. ZFYVE19 (zinc finger, FYVE domain containing 19), also known 1:100-1:1000), immunofluorescence (starting dilution 1:50, dilution range as MPFYVE (MLL partner containing FYVE domain), is a 471 amino acid pro - 1:50-1:500) and solid phase ELISA (starting dilution 1:30, dilution range 1:30- tein that contains one FYVE-type zinc finger. Expressed in heart, brain, kid ney, 1:3000); non cross-reactive with other ZFYVE family members. skeletal muscle and liver, ZFYVE19 may participate in transcriptional regula - Suitable for use as control antibody for ZFYVE19 siRNA (h): sc-90097, tion events within the cell. Defects in the gene encoding ZFYVE19 are asso - ZFYVE19 siRNA (m): sc-155604, ZFYVE19 shRNA Plasmid (h): sc-90097-SH, ciated with acute myeloblastic leukemia (AML), a rapidly progressing cancer ZFYVE19 shRNA Plasmid (m): sc-155604-SH, ZFYVE19 shRNA (h) Lentiviral of the myeloid line of white blood cells that is characterized by fever, ane mia, Particles: sc-90097-V and ZFYVE19 shRNA (m) Lentiviral Particles: bone pain, shortness of breath and frequent infections. Three isoforms of sc-155604-V. ZFYVE19 exist due to alternative splicing events. Molecular Weight of ZFYVE19: 48 kDa. REFERENCES Positive Controls: PLC/PRF/5 whole cell lysate.
    [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]