The USH1C 216G a Mutation and the 9-Repeat VNTR(T,T) Allele Are in Complete Linkage Disequilibrium in the Acadian Population

Total Page:16

File Type:pdf, Size:1020Kb

The USH1C 216G a Mutation and the 9-Repeat VNTR(T,T) Allele Are in Complete Linkage Disequilibrium in the Acadian Population Human Genetics DOI 10.1007/s00439-001-0653-7 Short Report The USH1C 216G A mutation and the 9-repeat VNTR(t,t) allele are in complete linkage disequilibrium in the Acadian population Sevtap Savas · Ben Frischhertz · Mary Z. Pelias · Mark A. Batzer · Prescott L. Deininger · Bronya J. B. Keats( ) S. Savas · M.Z. Pelias · B.J.B. Keats Department of Genetics, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA 70112, USA B. Frischhertz · P.L. Deininger Tulane Cancer Center, Department of Environmental Health Sciences, New Orleans, La., USA M.A. Batzer Department of Biological Sciences, Biological Computation and Visualization Center, Louisiana State University, Baton Rouge, La., USA E-mail: [email protected] Phone: +1-504-5686150 Fax: +1-504-5688500 Received: 25 May 2001 / Accepted: 31 October 2001 / Published online: Electronic database information: URLs for the data in this article are as follows: Primer3 software, http://www-genome.wi.mit.edu/cgi-bin/primer/primer3_www.cgi Abstract. Recently, mutations in USH1C were shown to be associated with Usher syndrome type IC, and a mutation (216G A) in exon 3 was identified in an Acadian family. In addition, a 45-bp variable number of tandem repeat (VNTR) polymorphism was found in intron 5 of USH1C. Polymerase chain reaction amplification of the VNTR region and restriction enzyme analysis of exon 3 of USH1C showed that, of 44 Acadian patients, 43 were homozygous for both the 216G A mutation and nine repeats of the VNTR, with a "t" nucleotide replacing a "g" nucleotide at the 8th position of both the eighth and ninth copies of the repeat, viz., 9VNTR(t,t). The remaining Acadian patient was reported to be a compound heterozygote for 216G A/9VNTR(t,t) and 238-239insC, a USH1C mutation that has been found in other populations. These data demonstrate that the 9VNTR(t,t) allele is in complete linkage disequilibrium with the 216G A mutation in the Acadian - 1 - population. Among 82 Acadian controls, one was heterozygous for 216G A/9VNTR(t,t). The 238-239insC mutation was not found in Acadian controls. Analysis of 340 non-Acadian normal samples showed the presence of a 9-repeat VNTR allele in one Hispanic sample. This individual had neither the 216G A mutation nor the Acadian VNTR(t,t) structure. These results suggest that the 216G A mutation and the 9VNTR(t,t) allele are restricted to the Acadians and are in complete linkage disequilibrium. Introduction Usher syndrome type IC (USH1C), an autosomal recessive disorder characterized by profound hearing impairment, early onset retinitis pigmentosa, and vestibular dysfunction, was recently shown to be caused by mutations in USH1C, a gene encoding a PDZ domain-containing protein named harmonin (Bitner-Glindzicz et al. 2000; Verpy et al. 2000). In Acadian USH1C patients, two molecular events specific to the Acadian population were found: (1) the 216G A mutation in exon 3 (Bitner-Glindzicz et al. 2000) and (2) a 9-repeat VNTR allele with an unusual structure in intron 5 (Verpy et al. 2000). The VNTR was shown to have a "t" nucleotide in the 8th position of the last copy of the repeat and a "g" nucleotide at this position in the preceding copies (VNTR(t)). In contrast, the 9-repeat VNTR allele associated with USH1C in Acadians had a "t" at the 8th position in both the eighth and ninth copies of the repeat, viz., 9VNTR(t,t). A single Acadian patient was reported to be a compound heterozygote for the Acadian 216G A mutation and a second mutation (238-239insC) that has been found in other populations (Bitner-Glindzicz et al. 2000; Verpy et al. 2000; Zwaeneopel et al. 2001). In this study, we have analyzed the frequency and association of the USH1C 216G A mutation and 9VNTR(t,t) allele in Acadians and in non-Acadian populations. Methods and materials The VNTR locus was genotyped by using the following primers: VNTR-F: 5’-ACCTTTTCACGGGGATCA-3’, and VNTR-R: 5’-GATCGTCCGGATCAATGG-3’. Polymerase chain reaction (PCR) amplification and restriction digestion analysis of the 216G A mutation was performed as explained in Verpy et al. (2000) and Bitner-Glindzicz et al. (2000), respectively. To distinguish between the VNTR(t,t) and VNTR(t) alleles, we designed an allele-specific primer (Wu et al. 1989) and paired it with the VNTR-F primer in a PCR. The sequence of the allele-specific primer was: VNTR-ARMS: 5’-GGAGGGCGGAGGAGCAGGT-3’. Samples were screened for the 238-239insC mutation by PCR amplification (Verpy et al. 2000) followed by sequencing with the BigDye Terminator Cycle Sequencing Reaction Kit (PE Applied Biosystems, Foster City, Calif.) and analyzed on an ABI PRIZM 377 DNA Sequencer (PE Applied Biosystems). The primers were designed by using Primer3 software (Rozen et al. 1998). Results and discussion Of the 44 Acadian USH1C patients analyzed, 43 were homozygous for both the 216G A mutation and the 9VNTR(t,t) allele. The remaining Acadian patient was a compound heterozygote for the 238-239insC and 216G A mutations, as previously described by Verpy et al. (2000). This patient was also heterozygous for the 9VNTR(t,t) allele. The results for these 44 patients demonstrate complete linkage disequilibrium between the 216G A mutation and the 9VNTR(t,t) allele in the Acadian population. - 2 - Analysis of both the VNTR locus and the 216G A mutation in 126 unrelated controls (82 Acadian, 44 European) showed that one Acadian individual was heterozygous for 216G A/9VNTR(t,t). None of these samples had the 216G A mutation but not the 9VNTR(t,t) allele, or conversely, the 9VNTR(t,t) allele without the 216G A mutation. A subset (n=69) of the Acadian controls was also screened for the 238-239insC mutation, and none had this mutation. These results, together with our finding of only one copy of the 238-239insC mutation among Acadian patients, suggest that this mutation is rare in Acadians and was probably introduced into this population very recently. Among the 340 unrelated controls who came from various populations and who were genotyped for the VNTR locus, viz., 65 African, 39 African-American, 71 Asian, 117 European (the 44 stated above plus an additional 73), and 48 Hispanic, one Hispanic sample was heterozygous for a 9-repeat VNTR allele. This individual did not have the 216G A mutation, and allele-specific amplification demonstrated that the structure of the 9-repeat VNTR was not 9VNTR(t,t) as shown in Fig. 1. Thus, the 9-repeat VNTR allele is rare in populations around the world, and the 9VNTR(t,t) allele is solely found in Acadians. - 3 - Fig. 1. A Schematic representation of the amplification recovery mutation system (ARMS) applied in this study. For simplicity, only 9VNTR(t,t) and 3-repeat VNTR(t) structures are depicted in this scheme, where each box represents the 45-bp repeating unit of the VNTR in USH1C. Above the boxes, the 8th nucleotide (either a "g" or a "t") in the repeating unit is shown. Long and small arrows Amplification primers VNTR-F and VNTR-ARMS, respectively. This PCR reaction is expected to yield a 117-bp product from a template containing a VNTR(t) structure, and 162-bp and 117-bp products from the 9VNTR(t,t) Acadian allele. B Gel analysis of ARMS PCR product. Lane 1 Negative PCR control, lane 2 Hispanic sample, lane 3 Acadian USH1C carrier, lane 4 Acadian USH1C patient, lane 5 control sample, lane 6 molecular weight standard, arrow position of the 100-bp fragment in the molecular weight standard (1 kb Plus DNA ladder, Life Technologies, Md., USA). Note that since the 117-bp product is also amplified from the 9VNTR(t,t) Acadian USH1C allele, the 117-bp product predominates in quantity over the 162-bp product in carriers of this allele The haplotype at markers surrounding USH1C and the 216G A/9VNTR(t,t) allele shows significant linkage disequilibrium over more than 6 cM (Keats et al. 1994; Nouri et al. 1994), suggesting that both the 216G A mutation and the 9VNTR(t,t) allele arose fairly recently in the Acadian population. Taking this information together with the absence of any chromosomes with the 216G A mutation but not the 9VNTR(t,t) allele, or vice versa, we conclude that the two events occurred at about the same time. However, it remains to be clarified whether the Acadian-specific 216G A mutation and the 9VNTR(t,t) allele arose simultaneously, and also whether the 9VNTR(t,t) allele contributes to the USH1C phenotype in Acadian patients. Acknowledgements. We are grateful to the Acadian families for their participation in this study. We thank San San Ng for helping with sequencing analysis. This work was supported by grants from the Foundation Fighting Blindness and the Louisiana Board of Regents Health Excellence Fund. - 4 - References Bitner-Glindzicz M, Lindley KJ, Rutland P, Blaydon D, Smith VV, Milla PJ, Hussain K, Furth-Lavi J, Cosgrove KE, Shepherd RM, Barnes PD, O’Brien RE, Farndon PA, Sowden J, Liu X-Z, Scanlan MJ, Malcolm S, Dunne MJ, Aynsley-Green A, Glaser B (2000) A recessive contiguous gene deletion syndrome causing infantile hyperinsulinism, enteropathy and deafness identifies the Usher type 1C gene. Nat Genet 26:56-60 Keats BJB, Nouri N, Pelias MZ, Deininger PL, Litt M (1994) Tightly linked flanking microsatellite markers for the Usher syndrome type I locus on the short arm of chromosome 11. Am J Hum Genet 54:681-686 Nouri N, Risch NJ, Pelias MZ, Litt M, Keats BJB (1994) Predicting the age of the mutation for Usher syndrome type I in the Acadian population.
Recommended publications
  • Comprehensive Sequence Analysis of Nine Usher Syndrome Genes in The
    Genotype-phenotype correlations J Med Genet: first published as 10.1136/jmedgenet-2011-100468 on 1 December 2011. Downloaded from ORIGINAL ARTICLE Comprehensive sequence analysis of nine Usher syndrome genes in the UK National Collaborative Usher Study Polona Le Quesne Stabej,1 Zubin Saihan,2,3 Nell Rangesh,4 Heather B Steele-Stallard,1 John Ambrose,5 Alison Coffey,5 Jenny Emmerson,5 Elene Haralambous,1 Yasmin Hughes,1 Karen P Steel,5 Linda M Luxon,4,6 Andrew R Webster,2,3 Maria Bitner-Glindzicz1,6 < Additional materials are ABSTRACT characterised by congenital, moderate to severe published online only. To view Background Usher syndrome (USH) is an autosomal hearing loss, with normal vestibular function and these files please visit the recessive disorder comprising retinitis pigmentosa, onset of RP around or after puberty; and type III journal online (http://jmg.bmj. fi com/content/49/1.toc). hearing loss and, in some cases, vestibular dysfunction. (USH3), de ned by postlingual progressive hearing 1 It is clinically and genetically heterogeneous with three loss and variable vestibular response together with Clinical and Molecular e 1 2 Genetics, Institute of Child distinctive clinical types (I III) and nine Usher genes RP. In addition there remain patients whose Health, UCL, London, UK identified. This study is a comprehensive clinical and disease does not fit into any of these three 2Institute of Ophthalmology, genetic analysis of 172 Usher patients and evaluates the subtypes, because of atypical audiovestibular or UCL, London, UK fi ‘ 3 contribution of digenic inheritance. retinal ndings, who are said to have atypical Moorfields Eye Hospital, Methods The genes MYO7A, USH1C, CDH23, PCDH15, ’ London, UK Usher syndrome .
    [Show full text]
  • Novel Mutations in the USH1C Gene in Usher Syndrome Patients
    Molecular Vision 2010; 16:2948-2954 <http://www.molvis.org/molvis/v16/a317> © 2010 Molecular Vision Received 30 September 2010 | Accepted 26 December 2010 | Published 31 December 2010 Novel mutations in the USH1C gene in Usher syndrome patients María José Aparisi,1 Gema García-García,1 Teresa Jaijo,1,2 Regina Rodrigo,1 Claudio Graziano,3 Marco Seri,3 Tulay Simsek,4 Enver Simsek,5 Sara Bernal,2,6 Montserrat Baiget,2,6 Herminio Pérez-Garrigues,2,7 Elena Aller,1,2 José María Millán1,2,8 1Grupo de Investigación en Enfermedades Neurosensoriales, Instituto de Investigación Sanitaria IIS-La Fe, Valencia, Spain; 2CIBER de Enfermedades Raras (CIBERER), Valencia, Spain; 3U.O. Genetica Medica, Policlinico S. Orsola-Malpighi, Università di Bologna, Italy; 4Ulucanlar Training and Research Eye Hospital, Ankara, Turkey; 5Department of Pediatric Endocrinology, Ankara Training and Research Hospital, Ankara, Turkey; 6Servicio de Genética, Hospital de la Santa Creu y Sant Pau. Barcelona, Spain; 7Servicio de Otorrinolaringología, Hospital Universitario La Fe, Valencia, Spain; 8Unidad de Genética y Diagnóstico Prenatal, Hospital Universitario La Fe, Valencia, Spain Purpose: Usher syndrome type I (USH1) is an autosomal recessive disorder characterized by severe-profound sensorineural hearing loss, retinitis pigmentosa, and vestibular areflexia. To date, five USH1 genes have been identified. One of these genes is Usher syndrome 1C (USH1C), which encodes a protein, harmonin, containing PDZ domains. The aim of the present work was the mutation screening of the USH1C gene in a cohort of 33 Usher syndrome patients, to identify the genetic cause of the disease and to determine the relative involvement of this gene in USH1 pathogenesis in the Spanish population.
    [Show full text]
  • NIDCD Fact Sheet Usher Syndrome Hearing Balance U.S
    NIDCD Fact Sheet Usher Syndrome hearing balance U.S. DEPARTMENT OF HEALTH & HUMAN SERVICES ∙ NATIONAL INSTITUTES OF HEALTH ∙ NATIONAL INSTITUTE ON DEAFNESS AND OTHER COMMUNICATION DISORDERS What is Usher syndrome? Who is affected by Usher syndrome? Usher syndrome is the most common condition that Approximately 3 to 6 percent of all children who are affects both hearing and vision. A syndrome is a deaf and another 3 to 6 percent of children who are disease or disorder that has more than one feature or hard-of-hearing have Usher syndrome. In developed symptom. The major symptoms of Usher syndrome countries such as the United States, about four babies are hearing loss and an eye disorder called retinitis in every 100,000 births have Usher syndrome. pigmentosa, or RP. RP causes night-blindness and a loss of peripheral vision (side vision) through the What causes Usher syndrome? progressive degeneration of the retina. The retina is Usher syndrome is inherited, which means that it is a light-sensitive tissue at the back of the eye and is passed from parents to their children through genes. crucial for vision (see photograph). As RP progresses, Genes are located in almost every cell of the body. the field of vision narrows—a condition known as Genes contain instructions that tell cells what to do. “tunnel vision”—until only central vision (the ability to Every person inherits two copies of each gene, one see straight ahead) remains. Many people with Usher from each parent. Sometimes genes are altered, syndrome also have severe balance problems. or mutated.
    [Show full text]
  • Deafblind Cajuns* by Phyllis Baudoin Grifard, Department of Biology, University of Louisiana at Lafayette
    NATIONAL CENTER FOR CASE STUDY TEACHING IN SCIENCE DeafBlind Cajuns* by Phyllis Baudoin Grifard, Department of Biology, University of Louisiana at Lafayette Part I – Meet Dan and Annie Annie pulled a chair for me at their dining room table and brought out snacks while her husband Dan made his way from the back of the house with his new diploma. Dan did not need to use his white cane inside their home. Since I am not fuent in American Sign Language (ASL), Annie ofered me a blank, boldly lined tablet labeled “low- vision notebook” and a thick black marker. I wrote “Congratulations!” when Dan proudly showed me his diploma from Gallaudet University (Figure 2), the only deaf-serving university in the world. He recently completed a BA degree in psychology and now is back home in Louisiana working as a case manager for deaf and deafblind residents at a local nursing home (Figure 3). Since Dan’s vision has deteriorated more than Annie’s, she used a tactile form of American Sign Language (tactile ASL) to convey what I wrote by signing into Dan’s hands. Figure 1. Dan and Annie. Dan and Annie are deafblind. Tey were born profoundly deaf and began to lose their peripheral and night vision in their late teens. Dan, now in his 50s, has only a small part of his central vision left. He uses adaptive technologies that magnify printed text and a white cane for mobility. Annie, who is in her 40s, has not lost as much of her peripheral vision yet, but navigating in low light, such as in dark restaurants, is becoming more difcult.
    [Show full text]
  • Mouse Models of Inherited Retinal Degeneration with Photoreceptor Cell Loss
    cells Review Mouse Models of Inherited Retinal Degeneration with Photoreceptor Cell Loss 1, 1, 1 1,2,3 1 Gayle B. Collin y, Navdeep Gogna y, Bo Chang , Nattaya Damkham , Jai Pinkney , Lillian F. Hyde 1, Lisa Stone 1 , Jürgen K. Naggert 1 , Patsy M. Nishina 1,* and Mark P. Krebs 1,* 1 The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; [email protected] (G.B.C.); [email protected] (N.G.); [email protected] (B.C.); [email protected] (N.D.); [email protected] (J.P.); [email protected] (L.F.H.); [email protected] (L.S.); [email protected] (J.K.N.) 2 Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand 3 Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand * Correspondence: [email protected] (P.M.N.); [email protected] (M.P.K.); Tel.: +1-207-2886-383 (P.M.N.); +1-207-2886-000 (M.P.K.) These authors contributed equally to this work. y Received: 29 February 2020; Accepted: 7 April 2020; Published: 10 April 2020 Abstract: Inherited retinal degeneration (RD) leads to the impairment or loss of vision in millions of individuals worldwide, most frequently due to the loss of photoreceptor (PR) cells. Animal models, particularly the laboratory mouse, have been used to understand the pathogenic mechanisms that underlie PR cell loss and to explore therapies that may prevent, delay, or reverse RD. Here, we reviewed entries in the Mouse Genome Informatics and PubMed databases to compile a comprehensive list of monogenic mouse models in which PR cell loss is demonstrated.
    [Show full text]
  • Analysis and Functional Evaluation of the Hair-Cell Transcriptome
    Analysis and functional evaluation of the hair-cell transcriptome Brian M. McDermott, Jr.*, Jessica M. Baucom, and A. J. Hudspeth† Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10021-6399 Contributed by A. J. Hudspeth, May 17, 2007 (sent for review March 31, 2007) An understanding of the molecular bases of the morphogenesis, ciated from the lagena, a receptor organ of the zebrafish’s ear. organization, and functioning of hair cells requires that the genes Linear amplification of the RNA from 200 hair cells yielded Ϸ40 expressed in these cells be identified and their functions ascer- ␮g of aRNA, an enhancement of Ϸ1 millionfold. The resultant tained. After purifying zebrafish hair cells and detecting mRNAs labeled aRNA was hybridized to an Affymetrix microarray with oligonucleotide microarrays, we developed a subtractive (Affymetrix, Santa Clara, CA) containing Ϸ15,000 oligonucle- strategy that identified 1,037 hair cell-expressed genes whose otide probe sets. Averaging the outcomes of three experiments cognate proteins subserve functions including membrane trans- (SI Data Set 1) resulted in the identification of 6,472 transcripts port, synaptic transmission, transcriptional control, cellular adhe- scored as ‘‘present’’ (SI Data Set 2). sion and signal transduction, and cytoskeletal organization. To In the second step, we defined the transcriptome from cells of assess the validity of the subtracted hair-cell data set, we verified a nonsensory organ, the liver (SI Data Set 1). Hepatocytes were the presence of 11 transcripts in inner-ear tissue. Functional eval- selected for the subtraction process for three reasons: they are uation of two genes from the subtracted data set revealed their nonneuronal and thus unlikely to express synaptic factors; they importance in hair bundles: zebrafish larvae bearing the seahorse lack cilia (http://members.global2000.net/bowser/cilialist.html) and ift 172 mutations display specific kinociliary defects.
    [Show full text]
  • A Novel Heterozygous Missense Variant (C.667G>T;P
    Original Article Diagnostic Genetics CROSSMARK_logo_3_Test 1 / 1 Ann Lab Med 2020;40:224-231 https://doi.org/10.3343/alm.2020.40.3.224 ISSN 2234-3806 • eISSN 2234-3814 https://crossmark-cdn.crossref.org/widget/v2.0/logos/CROSSMARK_Color_square.svg 2017-03-16 A Novel Heterozygous Missense Variant (c.667G>T;p. Gly223Cys) in USH1C That Interferes With Cadherin- Related 23 and Harmonin Interaction Causes Autosomal Dominant Nonsyndromic Hearing Loss Ju Sun Song , M.D.1,*, Amel Bahloul , Ph.D.2,3,4,5,*, Christine Petit , M.D., Ph.D.2,3,4,6, Sang Jin Kim , M.D., Ph.D.7, Il Joon Moon , M.D., Ph.D.8, Jinhyuk Lee , Ph.D.9,10, and Change-Seok Ki , M.D., Ph.D.1 1GC Genome, Yongin, Korea; 2Unité de génétique et physiologie de l’audition, Institut Pasteur, Paris, France; 3UMRS 1120, Inserm, Paris, France; 4Sorbonne Universités, Paris, France; 5Department of Otolaryngology - Head and Neck Surgery, Stanford University, Stanford, California, USA; 6College de France and Institut Pasteur, Paris, France; 7Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; 8Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; 9Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea; 10Department of Bioinformatics, University of Sciences and Technology, Daejeon, Korea Background: Pathogenic variants of USH1C, encoding a PDZ-domain-containing protein Received: June 13, 2019 called harmonin, have been known to cause autosomal recessive syndromic or nonsyn- Revision received: August 23, 2019 Accepted: November 26, 2019 dromic hearing loss (NSHL).
    [Show full text]
  • Download the Presentation Slides
    Usher Syndrome Type 1C Research Update Jennifer J. Lentz, PhD LSU Health-New Orleans, Louisiana Outline • Usher syndrome review • Characteristics and Prevalence • Types and subtypes • Clinical management and therapies under investigation • Lentz Lab Mission • USH1C gene • Knock-in mouse model • Antisense Therapy for Acadian USH1C • Targeting 216A mutation • Treatment of USH1C mice with ASOs • Gene Therapy for all USH1C • USH1C gene therapy development • Treatment of USH1C mice with gene replacement therapy Usher syndrome review – Prevalence and Types • Usher syndrome (USH or US) is the leading genetic cause of concurrent hearing and vision impairment. Some individuals also have imbalance. • Estimated 1 in ~20,000 individuals in the world have Usher • Currently, there are 3 clinical types and 11 subtypes (genes): Types: Type 1 (USH1) Type 2 (USH2) Type 3 (USH3) USH1B (MYO7A) USH2A (USH2A) USH3A (CLRN1) Subtypes: USH1C (USH1C) USH2C (ADGRV1) USH3B (HARS) USH1D (CDH23) USH2D (WHRN) USH1F (PCDH15) USH1G (SANS) USH1J (CIB2) Usher syndrome review - Diagnosis • Diagnosis is established with clinical features based on- • Severity of sensorineural hearing impairment (HI) • Presence of vestibular areflexia (imbalance) • Age of onset of retinitis pigmentosa (RP) – progressive visual loss that begins with night-blindness Type 1 (USH1) Type 2 (USH2) Type 3 (USH3) Severe-profound HI Mild - severe HI Post-lingual HI Vestibular areflexia RP beginning in late adolescence Variable Balance RP beginning in early adolescence RP beginning in adulthood Usher
    [Show full text]
  • PDZD7-MYO7A Complex Identified in Enriched Stereocilia Membranes
    RESEARCH ARTICLE PDZD7-MYO7A complex identified in enriched stereocilia membranes Clive P Morgan1†, Jocelyn F Krey1†, M’hamed Grati2, Bo Zhao3, Shannon Fallen1, Abhiraami Kannan-Sundhari2, Xue Zhong Liu2, Dongseok Choi4,5, Ulrich Mu¨ ller3, Peter G Barr-Gillespie1* 1Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, United States; 2Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, United States; 3Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, United States; 4OHSU-PSU School of Public Health, Oregon Health and Science University, Portland, United States; 5Graduate School of Dentistry, Kyung Hee University, Seoul, Korea Abstract While more than 70 genes have been linked to deafness, most of which are expressed in mechanosensory hair cells of the inner ear, a challenge has been to link these genes into molecular pathways. One example is Myo7a (myosin VIIA), in which deafness mutations affect the development and function of the mechanically sensitive stereocilia of hair cells. We describe here a procedure for the isolation of low-abundance protein complexes from stereocilia membrane fractions. Using this procedure, combined with identification and quantitation of proteins with mass spectrometry, we demonstrate that MYO7A forms a complex with PDZD7, a paralog of USH1C and DFNB31. MYO7A and PDZD7 interact in tissue-culture cells, and co-localize to the ankle-link region of stereocilia in wild-type but not Myo7a mutant mice. Our data thus describe a new paradigm for *For correspondence: gillespp@ the interrogation of low-abundance protein complexes in hair cell stereocilia and establish an ohsu.edu unanticipated link between MYO7A and PDZD7.
    [Show full text]
  • A Novel Locus for Usher Syndrome Type II, USH2B, Maps to Chromosome 3 at P23–24.2
    European Journal of Human Genetics (1999) 7, 363–367 © 1999 Stockton Press All rights reserved 1018–4813/99 $12.00 t http://www.stockton-press.co.uk/ejhg ARTICLE A novel locus for Usher syndrome type II, USH2B, maps to chromosome 3 at p23–24.2 Mounira Hmani1, Abdelmonem Ghorbel2, Amel Boulila-Elgaied1, Zeineb Ben Zina3, Wafa Kammoun2, Mohamed Drira2, Mohamed Chaabouni3, Christine Petit4 and Hammadi Ayadi1 1Laboratoire d’Immunologie et de Biologie Mol´eculaire, Facult´e de M´edecine, Sfax 2Service D’ORL, CHU Bourguiba, Sfax 3Service d’Ophtalmologie, CHU Bourguiba, Sfax, Tunisia 4Unit´e de G´en´etique des D´eficits Sensoriels, CNRS URA 1968, Institut Pasteur, Paris, France Usher type II syndrome is defined by the association of retinitis pigmentosa, appearing in the late second to early third decade of life, with congenital moderate to severe non-progressive hearing loss. This double sensory impairment is not accompanied by vestibular dysfunction. To date, only one Usher type II locus, USH2A, at chromosome band 1q41, has been defined. Here, we demonstrate by linkage analysis, that the gene responsible for Usher type II syndrome in a Tunisian consanguineous family maps to chromosome 3 at position p23–24.2, thus providing definitive evidence for the genetic heterogeneity of the syndrome. A maximum lod score of 4.3 was obtained with the polymorphic microsatellite markers corresponding to loci D3S1578, D3S3647 and D3S3658. This maps the gene underlying USH2B to a chromosomal region which overlaps the interval defined for the non-syndromic sensorineural recessive deafness DFNB6, raising the possibility that a single gene underlies both defects.
    [Show full text]
  • An Innovative Strategy for the Molecular Diagnosis of Usher Syndrome Identifies Causal Biallelic Mutations in 93% of European Patients
    European Journal of Human Genetics (2016) 24, 1730–1738 Official journal of The European Society of Human Genetics www.nature.com/ejhg ARTICLE An innovative strategy for the molecular diagnosis of Usher syndrome identifies causal biallelic mutations in 93% of European patients Crystel Bonnet1,2, Zied Riahi1,2, Sandra Chantot-Bastaraud3,4, Luce Smagghe1,2, Mélanie Letexier5, Charles Marcaillou5, Gaëlle M Lefèvre1,2, Jean-Pierre Hardelin6, Aziz El-Amraoui6, Amrit Singh-Estivalet1,2, Saddek Mohand-Saïd2,7,8, Susanne Kohl9, Anne Kurtenbach9, Ieva Sliesoraityte8,9, Ditta Zobor9, Souad Gherbi10, Francesco Testa11, Francesca Simonelli11, Sandro Banfi12,13, Ana Fakin14, Damjan Glavač15, Martina Jarc-Vidmar14, Andrej Zupan15, Saba Battelino16, Loreto Martorell Sampol17,MariaAntoniaClaveria17, Jaume Catala Mora17, Shzeena Dad18,LisbethBMøller18, Jesus Rodriguez Jorge17,MarkoHawlina14, Alberto Auricchio12,19, José-Alain Sahel2,7,8, Sandrine Marlin10, Eberhart Zrenner9,20, Isabelle Audo2,7,8 and Christine Petit*,1,2,6,21 Usher syndrome (USH), the most prevalent cause of hereditary deafness–blindness, is an autosomal recessive and genetically heterogeneous disorder. Three clinical subtypes (USH1–3) are distinguishable based on the severity of the sensorineural hearing impairment, the presence or absence of vestibular dysfunction, and the age of onset of the retinitis pigmentosa. A total of 10 causal genes, 6 for USH1, 3 for USH2, and 1 for USH3, and an USH2 modifier gene, have been identified. A robust molecular diagnosis is required not only to improve genetic counseling, but also to advance gene therapy in USH patients. Here, we present an improved diagnostic strategy that is both cost- and time-effective. It relies on the sequential use of three different techniques to analyze selected genomic regions: targeted exome sequencing, comparative genome hybridization, and quantitative exon amplification.
    [Show full text]
  • Target Sequencing of 307 Deafness Genes Identifies Candidate Genes Implicated in Microtia
    www.impactjournals.com/oncotarget/ Oncotarget, 2017, Vol. 8, (No. 38), pp: 63324-63332 Research Paper Target sequencing of 307 deafness genes identifies candidate genes implicated in microtia Pu Wang1, Xinmiao Fan1, Yibei Wang1, Yue Fan1, Yaping Liu2, Shuyang Zhang3 and Xiaowei Chen1 1Department of Otolaryngology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China 2Department of Medical Genetics, School of Basic Medicine, Peking Union Medical College, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China 3Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China Correspondence to: Xiaowei Chen, email: [email protected] Shuyang Zhang, email: [email protected] Keywords: microtia, deafness genes, next-generation sequencing, SKAT Received: April 23, 2017 Accepted: May 29, 2017 Published: June 28, 2017 Copyright: Wang et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Microtia is a congenital malformation of the external ear caused by genetic and/or environmental factors. However, no causal genetic mutations have been identified in isolated microtia patients. In this study, we utilized targeted genomic capturing combined with next-generation sequencing to screen for mutations in 307 deafness genes in 32 microtia patients. Forty-two rare heterozygous mutations in 25 genes, including 22 novel mutations in 24 isolated unilateral microtia cases were identified. Pathway analysis found five pathways especially focal adhesion pathway and ECM-receptor interaction pathway were significantly associated with microtia.
    [Show full text]