515 results on '"Usher Syndromes genetics"'
Search Results
202. Novel digenic inheritance of PCDH15 and USH1G underlies profound non-syndromic hearing impairment.
- Author
-
Schrauwen I, Chakchouk I, Acharya A, Liaqat K, Irfanullah, Nickerson DA, Bamshad MJ, Shah K, Ahmad W, and Leal SM
- Subjects
- Adult, Animals, Cadherin Related Proteins, Heterozygote, Humans, Male, Mechanotransduction, Cellular genetics, Multifactorial Inheritance genetics, Mutation genetics, Pakistan, Pedigree, Usher Syndromes genetics, Young Adult, Cadherins genetics, Hearing Loss genetics, Nerve Tissue Proteins genetics
- Abstract
Background: Digenic inheritance is the simplest model of oligenic disease. It can be observed when there is a strong epistatic interaction between two loci. For both syndromic and non-syndromic hearing impairment, several forms of digenic inheritance have been reported., Methods: We performed exome sequencing in a Pakistani family with profound non-syndromic hereditary hearing impairment to identify the genetic cause of disease., Results: We found that this family displays digenic inheritance for two trans heterozygous missense mutations, one in PCDH15 [p.(Arg1034His)] and another in USH1G [p.(Asp365Asn)]. Both of these genes are known to cause autosomal recessive non-syndromic hearing impairment and Usher syndrome. The protein products of PCDH15 and USH1G function together at the stereocilia tips in the hair cells and are necessary for proper mechanotransduction. Epistasis between Pcdh15 and Ush1G has been previously reported in digenic heterozygous mice. The digenic mice displayed a significant decrease in hearing compared to age-matched heterozygous animals. Until now no human examples have been reported., Conclusions: The discovery of novel digenic inheritance mechanisms in hereditary hearing impairment will aid in understanding the interaction between defective proteins and further define inner ear function and its interactome.
- Published
- 2018
- Full Text
- View/download PDF
203. Unravelling the pathogenic role and genotype-phenotype correlation of the USH2A p.(Cys759Phe) variant among Spanish families.
- Author
-
Pérez-Carro R, Blanco-Kelly F, Galbis-Martínez L, García-García G, Aller E, García-Sandoval B, Mínguez P, Corton M, Mahíllo-Fernández I, Martín-Mérida I, Avila-Fernández A, Millán JM, and Ayuso C
- Subjects
- Adult, Age of Onset, Aged, Amino Acid Substitution genetics, Blindness epidemiology, Blindness genetics, Genetic Association Studies, Heterozygote, Homozygote, Humans, Kaplan-Meier Estimate, Middle Aged, Retinitis Pigmentosa epidemiology, Spain epidemiology, Usher Syndromes epidemiology, Usher Syndromes genetics, Extracellular Matrix Proteins genetics, Retinitis Pigmentosa genetics
- Abstract
Introduction: Mutations in USH2A cause both isolated Retinitis Pigmentosa (RP) and Usher syndrome (that implies RP and hearing impairment). One of the most frequent variants identified in this gene and among these patients is the p.(Cys759Phe) change. However, the pathogenic role of this allele has been questioned since it was found in homozygosity in two healthy siblings of a Spanish family. To assess the causative role of USH2A p.(Cys759Phe) in autosomal recessive RP (ARRP) and Usher syndrome type II (USH2) and to establish possible genotype-phenotype correlations associated with p.(Cys759Phe), we performed a comprehensive genetic and clinical study in patients suffering from any of the two above-mentioned diseases and carrying at least one p.(Cys759Phe) allele., Materials and Methods: Diagnosis was set according to previously reported protocols. Genetic analyses were performed by using classical molecular and Next-Generation Sequencing approaches. Probands of 57 unrelated families were molecularly studied and 63 patients belonging to these families were phenotypically evaluated., Results: Molecular analysis characterized 100% of the cases, identifying: 11 homozygous patients for USH2A p.(Cys759Phe), 42 compound heterozygous patients (12 of them with another missense USH2A pathogenic variant and 30 with a truncating USH2A variant), and 4 patients carrying the p.(Cys759Phe) allele and a pathogenic variant in another RP gene (PROM1, CNGB1 or RP1). No additional causative variants were identified in symptomatic homozygous patients. Statistical analysis of clinical differences between zygosity states yielded differences (p≤0.05) in age at diagnosis of RP and hypoacusis, and progression of visual field loss. Homozygosity of p.(Cys759Phe) and compound heterozygosity with another USH2A missense variant is associated with ARRP or ARRP plus late onset hypoacusis (OR = 20.62, CI = 95%, p = 0.041)., Conclusions: The present study supports the role of USH2A p.(Cys759Phe) in ARRP and USH2 pathogenesis, and demonstrates the clinical differences between different zygosity states. Phenotype-genotype correlations may guide the genetic characterization based upon specific clinical signs and may advise on the clinical management and prognosis based upon a specific genotype., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
204. A novel homozygous variant of GPR98 causes usher syndrome type IIC in a consanguineous Chinese family by next generation sequencing.
- Author
-
Wei C, Yang L, Cheng J, Imani S, Fu S, Lv H, Li Y, Chen R, Leung EL, and Fu J
- Subjects
- Adult, Consanguinity, Female, Genetic Predisposition to Disease, Humans, Male, Pedigree, Phenotype, Prognosis, Asian People genetics, Codon, Nonsense, High-Throughput Nucleotide Sequencing methods, Homozygote, Receptors, G-Protein-Coupled genetics, Usher Syndromes genetics, Usher Syndromes pathology
- Abstract
Background: Usher syndrome (USH) is a common heterogeneous retinopathy and a hearing loss (HL) syndrome. However, the gene causing Usher syndrome type IIC (USH2C) in a consanguineous Chinese pedigree is unknown., Methods: We performed targeted next-generation sequencing analysis and Sanger sequencing to explore the GPR98 mutations in a USH2C pedigree that included a 32-year-old male patient from a consanguineous marriage family. Western blot verified the nonsense mutation., Results: To identify disease-causing gene variants in a consanguineous Chinese pedigree with USH2C, DNA from proband was analyzed using targeted next generation sequencing (NGS). The patient was clinically documented as a possible USH2 by a comprehensive auditory and ophthalmology evaluation. We succeeded in identifying the deleterious, novel, and homologous variant, c.6912dupG (p.Leu2305Valfs*4), in the GPR98 gene (NM_032119.3) that contributes to the progression of USH2C. Variant detected by targeted NGS was then confirmed and co-segregation was conducted by direct Sanger sequencing. Western blot verified losing almost two-thirds of its amino acid residues, including partial Calx-beta, whole EPTP and 7TM-GPCRs at the C-terminus of GPR98. Furthermore, our results highlighted that this p.Leu2305Valfs*4 variant is most likely pathogenic due to a large deletion at the seven-transmembrane G protein-coupled receptors (7TM-GPCRs) domain in GPR98 protein, leading to significantly decreased functionality and complex stability., Conclusions: These findings characterized the novel disease causativeness variant in GPR98 and broaden mutation spectrums, which could predict the pathogenic progression of patient with USH2C, guide diagnosis and treatment of this disease; and provide genetic counseling and family planning for consanguineous marriage pedigree in developing countries, including China.
- Published
- 2018
- Full Text
- View/download PDF
205. Utility of whole exome sequencing in the diagnosis of Usher syndrome: Report of novel compound heterozygous MYO7A mutations.
- Author
-
Ramzan K, Al-Owain M, Huma R, Al-Hazzaa SAF, Al-Ageel S, Imtiaz F, and Al-Sayed M
- Subjects
- Child, Preschool, Deafness etiology, Diagnosis, Differential, Female, Heterozygote, Homozygote, Humans, Male, Mutation, Myosin VIIa, Pedigree, Saudi Arabia, Usher Syndromes diagnosis, Deafness genetics, Myosins genetics, Usher Syndromes genetics, Exome Sequencing methods
- Abstract
Next generation sequencing (NGS), such as targeted panel sequencing, whole-exome sequencing and whole-genome sequencing has led to an exponential increase of elucidated genetic causes in both rare diseases, and common but heterogeneous disorders. NGS is applied in both research and clinical settings, and the clinical exome sequencing (CES), which provides not only the sequence variation data but also clinical interpretation, aids in reaching a final conclusion with regards to a genetic diagnosis. Usher syndrome is a group of disorders, characterized by bilateral sensorineural hearing loss, with or without vestibular dysfunction and retinitis pigmentosa. The index patient, a 2-year-old child was initially diagnosed with nonsyndromic hearing impairment. Homozygosity mapping followed by CES was utilized as a diagnostic tool to identify the genetic basis of his hearing loss. A paternally inherited novel insertion, c.198_199insA (p.Val67Serfs*73) and a maternally inherited novel deletion, c.1219_1226del (p.Phe407Aspfs*33) in gene MYO7A were found in compound heterozygous state in the index patient. The result expands the mutational spectrum of MYO7A. In addition it helped in early diagnosis of the syndrome, for planning and adjustments for the patient, and as well as for future family planning. This study highlights the clinical effectiveness of CES for Usher syndrome diagnosis in a child presented with congenital hearing loss., (Copyright © 2018. Published by Elsevier B.V.)
- Published
- 2018
- Full Text
- View/download PDF
206. Variants in CIB2 cause DFNB48 and not USH1J.
- Author
-
Booth KT, Kahrizi K, Babanejad M, Daghagh H, Bademci G, Arzhangi S, Zareabdollahi D, Duman D, El-Amraoui A, Tekin M, Najmabadi H, Azaiez H, and Smith RJ
- Subjects
- Adult, Female, Frameshift Mutation genetics, Genetic Linkage, Hearing Loss, Sensorineural diagnosis, Hearing Loss, Sensorineural physiopathology, Humans, Loss of Function Mutation genetics, Male, Middle Aged, Pedigree, Usher Syndromes diagnosis, Usher Syndromes physiopathology, Calcium-Binding Proteins genetics, Genetic Predisposition to Disease, Hearing Loss, Sensorineural genetics, Usher Syndromes genetics
- Abstract
The genetic, mutational and phenotypic spectrum of deafness-causing genes shows great diversity and pleiotropy. The best examples are the group of genes, which when mutated can either cause non-syndromic hearing loss (NSHL) or the most common dual sensory impairment, Usher syndrome (USH). Variants in the CIB2 gene have been previously reported to cause hearing loss at the DFNB48 locus and deaf-blindness at the USH1J locus. In this study, we characterize the phenotypic spectrum in a multiethnic cohort with autosomal recessive non-syndromic hearing loss (ARNSHL) due to variants in the CIB2 gene. Of the 6 families we ascertained, 3 segregated novel loss-of-function (LOF) variants, 2 families segregated missense variants (1 novel) and 1 family segregated a previously reported pathogenic variant in trans with a frameshift variant. This report is the first to show that biallelic LOF variants in CIB2 cause ARNSHL and not USH. In the era of precision medicine, providing the correct diagnosis (NSHL vs USH) is essential for patient care as it impacts potential intervention and prevention options for patients. Here, we provide evidence disqualifying CIB2 as an USH-causing gene., (© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
207. [Ciliopathies].
- Author
-
Gerth-Kahlert C and Koller S
- Subjects
- Abnormalities, Multiple diagnosis, Abnormalities, Multiple genetics, Abnormalities, Multiple physiopathology, Animals, Antigens, Neoplasm genetics, Bardet-Biedl Syndrome diagnosis, Bardet-Biedl Syndrome genetics, Bardet-Biedl Syndrome physiopathology, Cell Cycle Proteins, Cerebellum abnormalities, Cerebellum physiopathology, Cilia physiology, Ciliopathies genetics, Ciliopathies physiopathology, Cytoskeletal Proteins, DNA Mutational Analysis, Diagnosis, Differential, Disease Models, Animal, Eye Abnormalities diagnosis, Eye Abnormalities genetics, Eye Abnormalities physiopathology, Eye Proteins genetics, Genetic Association Studies, Genetic Diseases, X-Linked diagnosis, Genetic Diseases, X-Linked genetics, Genetic Diseases, X-Linked physiopathology, Genotype, Humans, Kidney Diseases, Cystic diagnosis, Kidney Diseases, Cystic genetics, Kidney Diseases, Cystic physiopathology, Leber Congenital Amaurosis diagnosis, Leber Congenital Amaurosis genetics, Leber Congenital Amaurosis physiopathology, Mice, Microtubule-Associated Proteins genetics, Myosin VIIa, Myosins genetics, Neoplasm Proteins genetics, Optic Atrophies, Hereditary diagnosis, Optic Atrophies, Hereditary genetics, Optic Atrophies, Hereditary physiopathology, Proteins genetics, Retina abnormalities, Retina physiopathology, Retinal Dystrophies genetics, Retinal Dystrophies physiopathology, Retinitis Pigmentosa diagnosis, Retinitis Pigmentosa genetics, Retinitis Pigmentosa physiopathology, Usher Syndromes diagnosis, Usher Syndromes genetics, Usher Syndromes physiopathology, Ciliopathies diagnosis, Retinal Dystrophies diagnosis
- Abstract
Ciliopathies are disorders caused by ciliary dysfunction and can affect an organ system or tissues. Isolated or syndromic retinal dystrophies are the most common ocular manifestation of ciliopathies. The photoreceptor connecting cilium plays a leading role in these ciliopathy-related retinal dystrophies. Dysfunctional photoreceptor cilia cause the most severe type of retinal dystrophy: Leber's congenital amaurosis (LCA). The most common syndromic ciliopathies with an ocular manifestation are Bardet-Biedl syndrome (BBS) and Usher syndrome. Molecular-genetic analysis revealed a large number of cilia genes with a high phenotype heterogeneity. Diagnosis of ciliopathies require a multi-disciplinary approach. Causative treatment of ciliopathies is not yet available; therefore, rehabilitative and supportive treatment is mandatory., Competing Interests: Die Autoren geben an, dass kein Interessenkonflikt besteht., (Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2018
- Full Text
- View/download PDF
208. Comprehensive Molecular Screening in Chinese Usher Syndrome Patients.
- Author
-
Sun T, Xu K, Ren Y, Xie Y, Zhang X, Tian L, and Li Y
- Subjects
- Adolescent, Adult, Analysis of Variance, Asian People genetics, Child, Child, Preschool, China, Female, Genetic Testing, Humans, Infant, Male, Middle Aged, Usher Syndromes ethnology, Usher Syndromes genetics, Young Adult, DNA Mutational Analysis methods, Molecular Diagnostic Techniques methods, Mutation, Usher Syndromes diagnosis
- Abstract
Purpose: Usher syndrome (USH) refers to a group of autosomal recessive disorders causing deafness and blindness. The objectives of this study were to determine the mutation spectrum in a cohort of Chinese patients with USH and to describe the clinical features of the patients with mutations., Methods: A total of 119 probands who were clinically diagnosed with USH were recruited for genetic analysis. All probands underwent ophthalmic examinations. A combination of molecular screening methods, including targeted next-generation sequencing, Sanger-DNA sequencing, and multiplex ligation probe amplification assay, was used to detect mutations., Results: We found biallelic mutations in 92 probands (77.3%), monoallelic mutations in 5 patients (4.2%), and 1 hemizygous mutation in 1 patient (0.8%), resulting in an overall mutation detection rate of 78.2%. Overall, 132 distinct disease-causing mutations involving seven USH (ABHD12, CDH23, GPR98, MYO7A, PCDH15, USH1C, and USH2A) genes; 5 other retinal degeneration genes (CHM, CNGA1, EYS, PDE6B, and TULP1); and 1 nonsyndromic hearing loss gene (MYO15A) were identified, and 78 were novel. Mutations of MYOA7 were responsible for 60% of USH1 families, followed by PCDH15 (20%) and USH1C (10%). Mutations of USH2A accounted for 67.7% of USH2 families, and mutation c.8559-2A>G was the most frequent one, accounting for 19.1% of the identified USH2A alleles., Conclusions: Our results confirm that the mutation spectrum for each USH gene in Chinese patients differs from those of other populations. The formation of the mutation profile for the Chinese population will enable a precise genetic diagnosis for USH patients in the future.
- Published
- 2018
- Full Text
- View/download PDF
209. [The Usher Syndrome, a Human Ciliopathy].
- Author
-
Wolfrum U and Nagel-Wolfrum K
- Subjects
- Animals, Ciliopathies classification, Ciliopathies genetics, Ciliopathies therapy, DNA Mutational Analysis, Deaf-Blind Disorders classification, Deaf-Blind Disorders diagnosis, Deaf-Blind Disorders genetics, Deaf-Blind Disorders therapy, Disease Models, Animal, Female, Humans, Infant, Newborn, Mice, Neonatal Screening, Photoreceptor Cells, Vertebrate physiology, Pregnancy, Usher Syndromes classification, Usher Syndromes genetics, Usher Syndromes therapy, Ciliopathies diagnosis, Rare Diseases, Usher Syndromes diagnosis
- Abstract
The human Usher syndrome (USH) is a complex, rare disease manifesting in its most common form of inherited deaf-blindness. Due to the heterogeneous manifestation of the clinical symptoms, three clinical types (USH1-3) are distinguished according to the severity of the disease pattern. For a correct diagnosis, in addition to the auditory tests in early newborn screening, ophthalmological examinations and molecular genetic analysis are important. Ten known USH genes encode proteins, which are from heterogeneous protein families, interact in functional protein networks. In the eye and in the ear, USH proteins are expressed primarily in the mechano-sensitive hair cells and the rod and cone photoreceptor cells, respectively. In the hair cells, the USH protein networks are essential for the correct differentiation of the hair bundles as well as for the function of the mechano-electrical transduction complex in the matured cell. In the photoreceptor cells, USH proteins are located in the ciliary region and participate in intracellular transport processes. In addition, a USH protein network is present in the so-called calyceal processes. The lack of calyceal processes and the absence of a prominent visual phenotype in the mouse disqualifies mice as models for studies on the ophthalmic component of USH. While hearing impairments can be compensated with hearing aids and cochlear implants, there is no practical therapy for USH in the eye. Currently, gene-based therapy concepts, such as gene addition, applications of antisense oligonucleotides and TRIDs ("translational readthrough inducing drugs") for the readthrough of nonsense mutations are preclinically evaluated. For USH1B/MYO7A the UshStat gene therapy clinical trial is ongoing., Competing Interests: Die Autoren geben an, dass kein Interessenkonflikt besteht., (Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2018
- Full Text
- View/download PDF
210. [Despite Challenges and Pitfalls: How Ophthalmology Benefits from the Use of Next-Generation Sequencing].
- Author
-
Bolz HJ
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Vesicular Transport, Alleles, Allelopathy genetics, Diagnosis, Differential, Eye Proteins genetics, Genetic Association Studies, Microtubule-Associated Proteins genetics, Retinal Dystrophies diagnosis, Retinal Dystrophies genetics, Sensitivity and Specificity, Tumor Suppressor Proteins genetics, Usher Syndromes diagnosis, Usher Syndromes genetics, Eye Diseases diagnosis, Eye Diseases genetics, High-Throughput Nucleotide Sequencing trends, Ophthalmology trends, Sequence Analysis, DNA trends
- Abstract
Within a few years, high-throughput sequencing (next-generation sequencing, NGS) has become a routine method in genetic diagnostics and has largely replaced conventional Sanger sequencing. The complexity of NGS data requires sound bioinformatic analysis: pinpointing the disease-causing variants may be difficult, and erroneous interpretations must be avoided. When looking at the group of retinal dystrophies as an example of eye disorders with extensive genetic heterogeneity, one can clearly say that NGS-based diagnostics yield important information for most patients and physicians, and that it has furthered our knowledge significantly. Furthermore, NGS has accelerated ophthalmogenetic research aimed at the identification of novel eye disease genes., Competing Interests: H. J. Bolz: bis Ende 2016 bei Bioscientia, einem Unternehmen medizinischer Diagnostik, angestellt. Auf den hier vorgelegten Artikel hatte dies keinen Einfluss, ein Interessenkonflikt besteht nicht., (Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2018
- Full Text
- View/download PDF
211. Antisense oligonucleotide therapy rescues disruptions in organization of exploratory movements associated with Usher syndrome type 1C in mice.
- Author
-
Donaldson TN, Jennings KT, Cherep LA, McNeela AM, Depreux FF, Jodelka FM, Hastings ML, and Wallace DG
- Subjects
- Animals, Behavior, Animal drug effects, Carrier Proteins metabolism, Cell Cycle Proteins, Cytoskeletal Proteins, Male, Mice, Usher Syndromes genetics, Usher Syndromes metabolism, Carrier Proteins genetics, Exploratory Behavior drug effects, Movement drug effects, Oligonucleotides, Antisense pharmacology, Usher Syndromes physiopathology
- Abstract
Usher syndrome, Type 1C (USH1C) is an autosomal recessive inherited disorder in which a mutation in the gene encoding harmonin is associated with multi-sensory deficits (i.e., auditory, vestibular, and visual). USH1C (Usher) mice, engineered with a human USH1C mutation, exhibit these multi-sensory deficits by circling behavior and lack of response to sound. Administration of an antisense oligonucleotide (ASO) therapeutic that corrects expression of the mutated USH1C gene, has been shown to increase harmonin levels, reduce circling behavior, and improve vestibular and auditory function. The current study evaluates the organization of exploratory movements to assess spatial organization in Usher mice and determine the efficacy of ASO therapy in attenuating any such deficits. Usher and heterozygous mice received the therapeutic ASO, ASO-29, or a control, non-specific ASO treatment at postnatal day five. Organization of exploratory movements was assessed under dark and light conditions at two and six-months of age. Disruptions in exploratory movement organization observed in control-treated Usher mice were consistent with impaired use of self-movement and environmental cues. In general, ASO-29 treatment rescued organization of exploratory movements at two and six-month testing points. These observations are consistent with ASO-29 rescuing processing of multiple sources of information and demonstrate the potential of ASO therapies to ameliorate topographical disorientation associated with other genetic disorders., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
212. The genetic dissection of Myo7a gene expression in the retinas of BXD mice.
- Author
-
Lu Y, Zhou D, King R, Zhu S, Simpson CL, Jones BC, Zhang W, Geisert EE, and Lu L
- Subjects
- Animals, Crosses, Genetic, Disease Models, Animal, Female, Gene Expression Profiling, Gene Expression Regulation, Gene Ontology, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Inbred DBA, Molecular Sequence Annotation, Myosin VIIa, Myosins metabolism, Polymorphism, Single Nucleotide, Signal Transduction, Transcription Factors metabolism, Usher Syndromes metabolism, Usher Syndromes pathology, Gene Regulatory Networks, Myosins genetics, Quantitative Trait Loci, Transcription Factors genetics, Usher Syndromes genetics
- Abstract
Purpose: Usher syndrome (US) is characterized by a loss of vision due to retinitis pigmentosa (RP) and deafness. US has three clinical subtypes, but even within each subtype, the severity varies. Myosin VIIA, coded by Myo7a , has been identified as one of the causal genes of US. This study aims to identify pathways and other genes through which Myo7a interacts to affect the presentation of US symptoms., Methods: In this study, we used the retinal tissue of BXD recombinant inbred (RI) mice to examine the expression of Myo7a and perform genetic mapping. Expression quantitative trait locus (eQTL), single nucleotide polymorphism (SNP), and gene correlation analysis were performed using GeneNetwork. Gene set enrichment analysis was performed using WebGestalt, and gene network construction was performed using the Gene Cohesion Analysis Tool., Results: We found Myo7a to be cis -regulated, with varied levels of expression across BXD strains. Here, we propose a genetic network with 40 genes whose expression is highly correlated with Myo7a . Among these genes, six have been linked to retinal diseases, three to deafness, and five share a transcription factor with Myo7a. Gene ontology and pathway analysis revealed a strong connection among ion channel activity, Myo7a , and US., Conclusions: Although Myo7a is a causal gene of US type I, this gene works with many other genes and pathways to affect the severity of US. Many of the genes found in the genetic network, pathways, and gene ontology categories of Myo7a are related to either deafness or blindness. Further investigation is needed to examine the specific relationships between these genes, which may assist in the treatment of US.
- Published
- 2018
213. Hearing impairment caused by mutations in two different genes responsible for nonsyndromic and syndromic hearing loss within a single family.
- Author
-
Niepokój K, Rygiel AM, Jurczak P, Kujko AA, Śniegórska D, Sawicka J, Grabarczyk A, Bal J, and Wertheim-Tysarowska K
- Subjects
- Child, Child, Preschool, Connexin 26, Connexins genetics, Extracellular Matrix Proteins genetics, Female, High-Throughput Nucleotide Sequencing, Humans, Male, Mutation, Pedigree, Deafness genetics, Usher Syndromes diagnosis, Usher Syndromes genetics
- Abstract
Usher syndrome is rare genetic disorder impairing two human senses, hearing and vision, with the characteristic late onset of vision loss. This syndrome is divided into three types. In all cases, the vision loss is postlingual, while loss of hearing is usually prelingual. The vestibular functions may also be disturbed in Usher type 1 and sometimes in type 3. Vestibular areflexia is helpful in making a proper diagnosis of the syndrome, but, often, the syndrome is misdiagnosed as a nonsyndromic hearing loss. Here, we present a Polish family with hearing loss, which was clinically classified as nonsyndromic. After excluding mutations in the DFNB1 locus, we implemented the next-generation sequencing method and revealed that hearing loss was syndromic and mutations in the USH2A gene indicate Usher syndrome. This research highlights the importance of molecular analysis in establishing a clinical diagnosis of congenital hearing loss.
- Published
- 2018
- Full Text
- View/download PDF
214. The Genetics of Usher Syndrome in the Israeli and Palestinian Populations.
- Author
-
Khalaileh A, Abu-Diab A, Ben-Yosef T, Raas-Rothschild A, Lerer I, Alswaiti Y, Chowers I, Banin E, Sharon D, and Khateb S
- Subjects
- Adult, Arabs, Child, Consanguinity, DNA Mutational Analysis, Female, Founder Effect, Genetic Testing, Genotype, Humans, Israel, Male, Myosin VIIa, Pedigree, Polymerase Chain Reaction, Usher Syndromes diagnosis, Young Adult, Ethnicity genetics, Extracellular Matrix Proteins genetics, Membrane Proteins genetics, Mutation, Myosins genetics, Polymorphism, Single Nucleotide, Usher Syndromes genetics
- Abstract
Purpose: Usher syndrome (USH) is the most common cause for deaf-blindness. It is genetically and clinically heterogeneous and prevalent in populations with high consanguinity rate. We aim to characterize the set of genes and mutations that cause USH in the Israeli and Palestinian populations., Methods: Seventy-four families with USH were recruited (23 with USH type 1 [USH1], 33 with USH2, seven with USH3, four with atypical USH, and seven families with an undetermined USH type). All affected subjects underwent a full ocular evaluation. A comprehensive genetic analysis, including Sanger sequencing for the detection of founder mutations, homozygosity mapping, and whole exome sequencing in large families was performed., Results: In 79% of the families (59 out of 74), an autosomal recessive inheritance pattern could be determined. Mutation detection analysis led to the identification of biallelic causative mutations in 51 (69%) of the families, including 21 families with mutations in USH2A, 17 in MYO7A, and seven in CLRN1. Our analysis revealed 28 mutations, 11 of which are novel (including c.802G>A, c.8558+1G>T, c.10211del, and c.14023A>T in USH2A; c.285+2T>G, c.2187+1G>T, c.3892G>A, c.5069_5070insC, c.5101C>T, and c.6196C>T in MYO7A; and c.15494del in GPR98)., Conclusions: We report here novel homozygous mutations in various genes causing USH, extending the spectrum of causative mutations. We also prove combined sequencing techniques as useful tools to identify novel disease-causing mutations. To the best of our knowledge, this is the largest report of a genetic analysis of Israeli and Palestinian families (n = 74) with different USH subtypes.
- Published
- 2018
- Full Text
- View/download PDF
215. A Natural Occurring Mouse Model with Adgrv1 Mutation of Usher Syndrome 2C and Characterization of its Recombinant Inbred Strains.
- Author
-
Yan W, Long P, Chen T, Liu W, Yao L, Ren Z, Li X, Wang J, Xue J, Tao Y, Zhang L, and Zhang Z
- Subjects
- Animals, Disease Models, Animal, Mice, Mutation, Usher Syndromes pathology, Exons, Inbreeding, Receptors, G-Protein-Coupled genetics, Usher Syndromes genetics
- Abstract
Background/aims: Our laboratory discovered a Kunming mouse with enormous electroretinogram (ERG) defects. Its auditory brainstem response (ABR) threshold was significantly elevated and closely resembled the features of Usher syndrome (USH). This study sought to cross these USH-like mice (named KMush/ush mice) with CBA/CaJ mice to establish recombinant inbred strains and identify their phenotypes and genotypes., Methods: KMush/ush mice were crossed with CBA/CaJ mice to establish inbred strains by sibling mating. ERG, ABR, ocular fundus morphology, histological examinations of the retina and inner ear, quantitative real-time polymerase chain reaction, western blotting, and exon sequencing were performed to assess the phenotypes and genotypes of the offspring strains., Results: The F1 hybrids from crossing KMush/ush and CBA/CaJ mice had normal ERG and ABR responses. The F2 offspring from intercrossing the F1 mice showed a segregation of the retinitis pigmentosa (RP) and hearing loss phenotypes. The CBA-1ush/ush mice had an RP phenotype that was characterized by a vanished ERG waveform and loss of the outer nuclear layer. Their Pde6b gene had a nonsense mutation that resulted in the failure of protein production in western blotting. However, the ABR threshold of this strain of mice was normal. The CBA-2ush/ush mice had normal retinal function and architecture. Their ABR threshold was increased, with a dramatic degeneration of the stereocilia bundles in the outer hair cells of the inner ear. Whole exome sequencing and exon sequencing revealed a deletion of one base pair in exon 31 of the Adgrv1 gene, which would result in the premature termination of protein encoding. The level of Adgrv1 mRNA was reduced in the CBA-2ush/ush mice. The CBA-3ush/ush mice had phenotypes of RP, elevated ABR threshold, and degeneration of the stereocilia bundles in the outer hair cells. They were closely associated with the nonsense mutations of Pde6b and Adgrv1, respectively., Conclusion: We isolated a mouse strain with hearing loss from inbred mice with retinal degeneration and established it as a recombinant inbred strain with a spontaneous mutation in Adgrv1, the human Usher syndrome 2C gene. The retinal degeneration was cause by a mutation in Pde6b, while the hearing loss was caused by a mutation in Adgrv1., (© 2018 The Author(s). Published by S. Karger AG, Basel.)
- Published
- 2018
- Full Text
- View/download PDF
216. Targeted next generation sequencing identified a novel mutation in MYO7A causing Usher syndrome type 1 in an Iranian consanguineous pedigree.
- Author
-
Kooshavar D, Razipour M, Movasat M, and Keramatipour M
- Subjects
- Codon, Nonsense, Consanguinity, Female, Homozygote, Humans, Iran, Male, Mutation, Myosin VIIa, Pedigree, Phenotype, Polymerase Chain Reaction, High-Throughput Nucleotide Sequencing methods, Myosins genetics, Usher Syndromes genetics
- Abstract
Background: Usher syndrome (USH) is characterized by congenital hearing loss and retinitis pigmentosa (RP) with a later onset. It is an autosomal recessive trait with clinical and genetic heterogeneity which makes the molecular diagnosis much difficult. In this study, we introduce a pedigree with two affected members with USH type 1 and represent a cost and time effective approach for genetic diagnosis of USH as a genetically heterogeneous disorder., Methods: Target region capture in the genes of interest, followed by next generation sequencing (NGS) was used to determine the causative mutations in one of the probands. Then segregation analysis in the pedigree was conducted using PCR-Sanger sequencing., Results: Targeted NGS detected a novel homozygous nonsense variant c.4513G > T (p.Glu1505Ter) in MYO7A. The variant is segregating in the pedigree with an autosomal recessive pattern., Conclusion: In this study, a novel stop gained variant c.4513G > T (p.Glu1505Ter) in MYO7A was found in an Iranian pedigree with two affected members with USH type 1. Bioinformatic as well as pedigree segregation analyses were in line with pathogenic nature of this variant. Targeted NGS panel was showed to be an efficient method for mutation detection in hereditary disorders with locus heterogeneity., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
217. Identification of two novel pathogenic compound heterozygous MYO7A mutations in Usher syndrome by whole exome sequencing.
- Author
-
Jia Y, Li X, Yang D, Xu Y, Guo Y, and Li X
- Subjects
- Adolescent, Child, Codon, Nonsense, Female, Heterozygote, Humans, Male, Mutation, Myosin VIIa, Pedigree, Polymerase Chain Reaction, Exome Sequencing methods, Myosins genetics, Usher Syndromes genetics
- Abstract
The current study aims to identify the pathogenic sites in a core pedigree of Usher syndrome (USH). A core pedigree of USH was analyzed by whole exome sequencing (WES). Mutations were verified by polymerase chain reaction (PCR) amplification and Sanger sequencing. Two pathogenic variations (c.849+2T>C and c.5994G>A) in MYO7A were successfully identified and individually separated from parents. One variant (c.849+2T>C) was nonsense mutation, causing the protein terminated in advance, and the other one (c.5994G>A) located near the boundary of exon could cause aberrant splicing. This study provides a meaningful exploration for identification of clinical core genetic pedigrees., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
218. Visual Outcomes in Japanese Patients with Retinitis Pigmentosa and Usher Syndrome Caused by USH2A Mutations.
- Author
-
Nagase Y, Kurata K, Hosono K, Suto K, Hikoya A, Nakanishi H, Mizuta K, Mineta H, Minoshima S, and Hotta Y
- Subjects
- Adolescent, Adult, Child, DNA Mutational Analysis, Exons, Extracellular Matrix Proteins metabolism, Female, Genotype, Humans, Incidence, Japan epidemiology, Male, Middle Aged, Pedigree, Prognosis, Retinitis Pigmentosa epidemiology, Retinitis Pigmentosa metabolism, Usher Syndromes epidemiology, Usher Syndromes metabolism, Young Adult, DNA genetics, Extracellular Matrix Proteins genetics, Mutation, Retinitis Pigmentosa genetics, Usher Syndromes genetics, Visual Acuity, Visual Fields physiology
- Abstract
Purpose: EYS and USH2A are the most common causative genes for retinitis pigmentosa (RP) in Japan. We determined the clinical outcomes for USH2A-related non-syndromic RP or Usher syndrome type II (USH2)., Methods: Two non-syndromic RP and 11 USH2 patients with previously identified USH2A mutations were included. Their complete history and medical records were collected using standard procedures. Visual fields and acuity were compared with those of patients with EYS mutations. Clinical analyses were based on ophthalmic and otolaryngologic examinations., Results: In all patients, the fundus displayed changes typical of RP. Most patients showed relatively well-preserved visual acuity in their thirties or forties, with rapid deterioration in their fifties. Concentric constriction started in the twenties or thirties, and no effective residual visual field was observed after the fifties., Conclusions: The visual outcome for non-syndromic RP or USH2 patients with USH2A mutations is consistent with that for RP patients with EYS mutations.
- Published
- 2018
- Full Text
- View/download PDF
219. Panel sequencing of 264 candidate susceptibility genes and segregation analysis in a cohort of non-BRCA1, non-BRCA2 breast cancer families.
- Author
-
Li J, Li H, Makunin I, Thompson BA, Tao K, Young EL, Lopez J, Camp NJ, Tavtigian SV, John EM, Andrulis IL, Khanna KK, Goldgar D, and Chenevix-Trench G
- Subjects
- Adult, BRCA1 Protein genetics, BRCA2 Protein genetics, Breast Neoplasms pathology, DNA-Binding Proteins genetics, Extracellular Matrix Proteins genetics, Female, Gene Expression Regulation, Neoplastic, Germ-Line Mutation, Humans, Middle Aged, Pedigree, Risk, Transcription Factors genetics, Usher Syndromes pathology, Breast Neoplasms genetics, Epigenesis, Genetic genetics, Genetic Predisposition to Disease, Usher Syndromes genetics
- Abstract
Purpose: The main aim of this study was to screen epigenetic modifier genes and known breast cancer driver genes for germline mutations in non-BRCA1/2 (BRCAx) breast cancer families in order to identify novel susceptibility genes of moderate-high penetrance., Methods: We screened 264 candidate susceptibility genes in 656 index cases from non-BRCA1/2 families. Potentially pathogenic candidate mutations were then genotyped in all available family members for the assessment of co-segregation of the variant with disease in the family in order to estimate the breast cancer risks associated with these mutations. For 11 of the candidate susceptibility genes, we screened an additional 800 non-BRCA1/2 breast cancer cases and 787 controls., Results: Only two genes, CHD8 and USH2A showed any evidence of an increased risk of breast cancer (RR = 2.40 (95% CI 1.0-7.32) and 2.48 (95% CI 1.11-6.67), respectively)., Conclusions: We found no convincing evidence that epigenetic modifier and known breast cancer driver genes carry germline mutations that increase breast cancer risk. USH2A is no longer regarded as a breast cancer driver gene and seems an implausible candidate given its association with Usher syndrome. However, somatic mutations in CHD8 have been recently reported, making it an even more promising candidate, but further analysis of CHD8 in very large cohorts of families or case-control studies would be required to determine if it is a moderate-risk breast cancer susceptibility gene.
- Published
- 2017
- Full Text
- View/download PDF
220. Targeted next generation sequencing in Italian patients with Usher syndrome: phenotype-genotype correlations.
- Author
-
Eandi CM, Dallorto L, Spinetta R, Micieli MP, Vanzetti M, Mariottini A, Passerini I, Torricelli F, Alovisi C, and Marchese C
- Subjects
- Adolescent, Adult, Child, DNA Mutational Analysis, Female, Genetic Association Studies, Genetic Predisposition to Disease, Genotype, High-Throughput Nucleotide Sequencing, Humans, Italy, Male, Middle Aged, Mutation, Missense genetics, Myosin VIIa, Pedigree, Sequence Deletion genetics, Usher Syndromes classification, Usher Syndromes pathology, Young Adult, Extracellular Matrix Proteins genetics, Myosins genetics, Receptors, G-Protein-Coupled genetics, Usher Syndromes genetics
- Abstract
We report results of DNA analysis with next generation sequencing (NGS) of 21 consecutive Italian patients from 17 unrelated families with clinical diagnosis of Usher syndrome (4 USH1 and 17 USH2) searching for mutations in 11 genes: MYO7A, CDH23, PCDH15, USH1C, USH1G, USH2A, ADGVR1, DFNB31, CLRN1, PDZD7, HARS. Likely causative mutations were found in all patients: 25 pathogenic variants, 18 previously reported and 7 novel, were identified in three genes (USH2A, MYO7A, ADGRV1). All USH1 presented biallelic MYO7A mutations, one USH2 exhibited ADGRV1 mutations, whereas 16 USH2 displayed USH2A mutations. USH1 patients experienced hearing problems very early in life, followed by visual impairment at 1, 4 and 6 years. Visual symptoms were noticed at age 20 in a patient with homozygous novel MYO7A missense mutation c.849G > A. USH2 patients' auditory symptoms, instead, arose between 11 months and 14 years, while visual impairment occurred later on. A homozygous c.5933_5940del;5950_5960dup in USH2A was detected in one patient with early deafness. One patient with homozygous deletion from exon 23 to 32 in USH2A suffered early visual symptoms. Therefore, the type of mutation in USH2A and MYO7A genes seems to affect the age at which both auditory and visual impairment occur in patients with USH.
- Published
- 2017
- Full Text
- View/download PDF
221. Integrin α8 and Pcdh15 act as a complex to regulate cilia biogenesis in sensory cells.
- Author
-
Goodman L and Zallocchi M
- Subjects
- Actin Cytoskeleton metabolism, Actin Cytoskeleton ultrastructure, Animals, Cadherin Related Proteins, Cadherins antagonists & inhibitors, Cadherins deficiency, Centrioles metabolism, Centrioles ultrastructure, Cilia ultrastructure, Disease Models, Animal, Endocytosis, Gene Expression Regulation, Developmental, Hair Cells, Auditory ultrastructure, Humans, Integrins antagonists & inhibitors, Integrins deficiency, Larva genetics, Larva growth & development, Larva metabolism, Monomeric GTP-Binding Proteins genetics, Monomeric GTP-Binding Proteins metabolism, Mutation, Protein Binding, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Signal Transduction, Usher Syndromes genetics, Usher Syndromes metabolism, Usher Syndromes pathology, Zebrafish growth & development, Zebrafish metabolism, Zebrafish Proteins antagonists & inhibitors, Zebrafish Proteins deficiency, Zebrafish Proteins metabolism, rab GTP-Binding Proteins genetics, rab GTP-Binding Proteins metabolism, Cadherins genetics, Cilia metabolism, Hair Cells, Auditory metabolism, Integrins genetics, Zebrafish genetics, Zebrafish Proteins genetics
- Abstract
The way an organism perceives its surroundings depends on sensory systems and the highly specialized cilia present in the neurosensory cells. Here, we describe the existence of an integrin α8 (Itga8) and protocadherin-15a (Pcdh15a) ciliary complex in neuromast hair cells in a zebrafish model. Depletion of the complex via downregulation or loss-of-function mutation leads to a dysregulation of cilia biogenesis and endocytosis. At the molecular level, removal of the complex blocks the access of Rab8a into the cilia as well as normal recruitment of ciliary cargo by centriolar satellites. These defects can be reversed by the introduction of a constitutively active form of Rhoa, suggesting that Itga8-Pcdh15a complex mediates its effect through the activation of this small GTPase and probably by the regulation of actin cytoskeleton dynamics. Our data points to a novel mechanism involved in the regulation of sensory cilia development, with the corresponding implications for normal sensory function., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2017. Published by The Company of Biologists Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
222. Usher's Syndrome Type II: A Comparative Study of Genetic Mutations and Vestibular System Evaluation.
- Author
-
Magliulo G, Iannella G, Gagliardi S, Iozzo N, Plateroti R, Mariottini A, and Torricelli F
- Subjects
- Adolescent, Adult, Cross-Sectional Studies, Female, Humans, Italy, Male, Middle Aged, Mutation, Vestibular Function Tests, Extracellular Matrix Proteins genetics, Receptors, G-Protein-Coupled genetics, Usher Syndromes genetics, Usher Syndromes physiopathology
- Abstract
Objective Usher's syndrome type II (USH2) is characterized by moderate to profound congenital hearing loss, later onset of retinitis pigmentosa, and normal vestibular function. Recently, a study investigating the vestibular function of USH2 patients demonstrated a pathologic response to vestibular tests. In this cross-sectional study we performed vestibular tests of a group patients with genetic diagnosis of USH2 syndrome to demonstrate if vestibular damage is present in USH2 patients. Study Design Cross-sectional study. Setting Tertiary referral center. Subjects and Methods Mutated genes of 7 patients with a clinical diagnosis of USH2 were evaluated. Vestibular function was investigated by audiometry, Fitzgerald-Hallpike caloric vestibular testing, cervical vestibular evoked myogenic potentials (C-VEMPs), ocular vestibular evoked myogenic potentials (O-VEMPs), and video head impulse test (v-HIT). Results Genetic tests confirmed the USH2 diagnosis in 5 of 7 patients examined, with 1 patient reporting a unique mutation on genetic tests. Four (80%) of the 5 patients with a genetic diagnosis of USH2 showed pathological O-VEMPs. Two patients (40%) reported bilateral absent or abnormal values of C-VEMPs. The superior semicircular canal presented a significant deficit in 2 (40%) patients. The same 2 cases showed a pathologic response of the v-HIT of the horizontal semicircular canal. Finally, the posterior semicircular canal presented a significant deficit in 4 (40.0%) patients. Conclusion A vestibular evaluation with vestibular evoked myogenic potentials and v-HIT seems to identify latent damage to the vestibular receptors of USH2 patients.
- Published
- 2017
- Full Text
- View/download PDF
223. Genetic analysis of a Chinese family with members affected with Usher syndrome type II and Waardenburg syndrome type IV.
- Author
-
Wang X, Lin XJ, Tang X, Chai YC, Yu DH, Chen DY, and Wu H
- Subjects
- Adult, Aged, Asian People genetics, Female, Genetic Testing, High-Throughput Nucleotide Sequencing, Hirschsprung Disease complications, Homozygote, Humans, Male, Middle Aged, Mutation, Pedigree, Phenotype, Polymerase Chain Reaction, Usher Syndromes complications, Waardenburg Syndrome complications, Hirschsprung Disease genetics, Usher Syndromes genetics, Waardenburg Syndrome genetics
- Abstract
Aims: The purpose of this study was to identify the genetic causes of a family presenting with multiple symptoms overlapping Usher syndrome type II (USH2) and Waardenburg syndrome type IV (WS4)., Methods: Targeted next-generation sequencing including the exon and flanking intron sequences of 79 deafness genes was performed on the proband. Co-segregation of the disease phenotype and the detected variants were confirmed in all family members by PCR amplification and Sanger sequencing., Results: The affected members of this family had two different recessive disorders, USH2 and WS4. By targeted next-generation sequencing, we identified that USH2 was caused by a novel missense mutation, p.V4907D in GPR98; whereas WS4 due to p.V185M in EDNRB. This is the first report of homozygous p.V185M mutation in EDNRB in patient with WS4., Conclusion: This study reported a Chinese family with multiple independent and overlapping phenotypes. In condition, molecular level analysis was efficient to identify the causative variant p.V4907D in GPR98 and p.V185M in EDNRB, also was helpful to confirm the clinical diagnosis of USH2 and WS4., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
224. A homozygous MYO7A mutation associated to Usher syndrome and unilateral auditory neuropathy spectrum disorder.
- Author
-
Xia H, Hu P, Yuan L, Xiong W, Xu H, Yi J, Yang Z, Deng X, Guo Y, and Deng H
- Subjects
- Asian People genetics, Audiometry, Base Sequence, DNA Mutational Analysis, Evoked Potentials, Auditory, Brain Stem, Family, Female, Hearing Loss, Central physiopathology, Homozygote, Humans, Male, Middle Aged, Myosin VIIa, Otoacoustic Emissions, Spontaneous, Pedigree, Usher Syndromes physiopathology, Exome Sequencing, Hearing Loss, Central genetics, Mutation genetics, Myosins genetics, Usher Syndromes genetics
- Abstract
Usher syndrome (USH) is an autosomal recessive disorder characterized by sensorineural hearing loss, progressive visual loss and night blindness due to retinitis pigmentosa (RP), with or without vestibular dysfunction. The purpose of this study was to detect the causative gene in a consanguineous Chinese family with USH. A c.3696_3706del (p.R1232Sfs*72) variant in the myosin VIIa gene (MYO7A) was identified in the homozygous state by exome sequencing. The co‑segregation of the MYO7A c.3696_3706del variant with the phenotype of deafness and progressive visual loss in the USH family was confirmed by Sanger sequencing. The variant was absent in 200 healthy controls. Therefore, the c.3696_3706del variant may disrupt the interaction between myosin VIIa and other USH1 proteins, and impair melanosome transport in retinal pigment epithelial cells. Notably, bilateral auditory brainstem responses were absent in two patients of the USH family, while distortion product otoacoustic emissions were elicited in the right ears of the two patients, consistent with clinical diagnosis of unilateral auditory neuropathy spectrum disorder. These data suggested that the homozygous c.3696_3706del variant in the MYO7A gene may be the disease‑causing mutation for the disorder in this family. These findings broaden the phenotype spectrum of the MYO7A gene, and may facilitate understanding of the molecular pathogenesis of the disease, and genetic counseling for the family.
- Published
- 2017
- Full Text
- View/download PDF
225. Novel compound heterozygous mutations in the GPR98 (USH2C) gene identified by whole exome sequencing in a Moroccan deaf family.
- Author
-
Bousfiha A, Bakhchane A, Charoute H, Detsouli M, Rouba H, Charif M, Lenaers G, and Barakat A
- Subjects
- Adolescent, Adult, Child, Female, Heterozygote, Humans, Male, Morocco, Mutation genetics, Pedigree, Siblings, Exome Sequencing methods, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Usher Syndromes genetics
- Abstract
In the present work, we identified two novel compound heterozygote mutations in the GPR98 (G protein-coupled receptor 98) gene causing Usher syndrome. Whole-exome sequencing was performed to study the genetic causes of Usher syndrome in a Moroccan family with three affected siblings. We identify two novel compound heterozygote mutations (c.1054C > A, c.16544delT) in the GPR98 gene in the three affected siblings carrying post-linguale bilateral moderate hearing loss with normal vestibular functions and before installing visual disturbances. This is the first time that mutations in the GPR98 gene are described in the Moroccan deaf patients.
- Published
- 2017
- Full Text
- View/download PDF
226. Rescue of peripheral vestibular function in Usher syndrome mice using a splice-switching antisense oligonucleotide.
- Author
-
Vijayakumar S, Depreux FF, Jodelka FM, Lentz JJ, Rigo F, Jones TA, and Hastings ML
- Subjects
- Animals, Cell Cycle Proteins, Cytoskeletal Proteins, Disease Models, Animal, Evoked Potentials, Auditory, Hearing genetics, Mice, Mutation, Oligonucleotides, Antisense therapeutic use, Retina metabolism, Retinal Degeneration genetics, Usher Syndromes genetics, Usher Syndromes metabolism, Vestibular Evoked Myogenic Potentials genetics, Vestibule, Labyrinth metabolism, Vestibule, Labyrinth physiology, Carrier Proteins genetics, Carrier Proteins metabolism, Usher Syndromes therapy
- Abstract
Usher syndrome type 1C (USH1C/harmonin) is associated with profound retinal, auditory and vestibular dysfunction. We have previously reported on an antisense oligonucleotide (ASO-29) that dramatically improves auditory function and balance behavior in mice homozygous for the harmonin mutation Ush1c c.216G > A following a single systemic administration. The findings were suggestive of improved vestibular function; however, no direct vestibular assessment was made. Here, we measured vestibular sensory evoked potentials (VsEPs) to directly assess vestibular function in Usher mice. We report that VsEPs are absent or abnormal in Usher mice, indicating profound loss of vestibular function. Strikingly, Usher mice receiving ASO-29 treatment have normal or elevated vestibular response thresholds when treated during a critical period between postnatal day 1 and 5, respectively. In contrast, treatment of mice with ASO-29 treatment at P15 was minimally effective at rescuing vestibular function. Interestingly, ASO-29 treatment at P1, P5 or P15 resulted in sufficient vestibular recovery to support normal balance behaviors, suggesting a therapeutic benefit to balance with ASO-29 treatment at P15 despite the profound vestibular functional deficits that persist with treatment at this later time. These findings provide the first direct evidence of an effective treatment of peripheral vestibular function in a mouse model of USH1C and reveal the potential for using antisense technology to treat vestibular dysfunction., (© The Author 2017. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
227. Local gene therapy durably restores vestibular function in a mouse model of Usher syndrome type 1G.
- Author
-
Emptoz A, Michel V, Lelli A, Akil O, Boutet de Monvel J, Lahlou G, Meyer A, Dupont T, Nouaille S, Ey E, Franca de Barros F, Beraneck M, Dulon D, Hardelin JP, Lustig L, Avan P, Petit C, and Safieddine S
- Subjects
- Animals, Animals, Newborn, DNA, Complementary administration & dosage, DNA, Complementary genetics, Dependovirus genetics, Disease Models, Animal, Evoked Potentials, Auditory, Brain Stem, Genetic Therapy methods, Genetic Vectors, Hair Cells, Auditory pathology, Hair Cells, Auditory physiology, Humans, Mice, Mice, Knockout, Microscopy, Electron, Scanning, Nerve Tissue Proteins deficiency, Nerve Tissue Proteins genetics, Usher Syndromes physiopathology, Vestibule, Labyrinth pathology, Vestibule, Labyrinth physiopathology, Usher Syndromes genetics, Usher Syndromes therapy
- Abstract
Our understanding of the mechanisms underlying inherited forms of inner ear deficits has considerably improved during the past 20 y, but we are still far from curative treatments. We investigated gene replacement as a strategy for restoring inner ear functions in a mouse model of Usher syndrome type 1G, characterized by congenital profound deafness and balance disorders. These mice lack the scaffold protein sans, which is involved both in the morphogenesis of the stereociliary bundle, the sensory antenna of inner ear hair cells, and in the mechanoelectrical transduction process. We show that a single delivery of the sans cDNA by the adenoassociated virus 8 to the inner ear of newborn mutant mice reestablishes the expression and targeting of the protein to the tips of stereocilia. The therapeutic gene restores the architecture and mechanosensitivity of stereociliary bundles, improves hearing thresholds, and durably rescues these mice from the balance defects. Our results open up new perspectives for efficient gene therapy of cochlear and vestibular disorders by showing that even severe dysmorphogenesis of stereociliary bundles can be corrected., Competing Interests: Conflict of interest statement: A patent involving A.E., C.P., and S.S. (PCT/EP2016/053613) has been deposited by the Institut Pasteur, INSERM, and CNRS.
- Published
- 2017
- Full Text
- View/download PDF
228. Pigmentary retinopathy, rod-cone dysfunction and sensorineural deafness associated with a rare mitochondrial tRNA Lys (m.8340G>A) gene variant.
- Author
-
Gill JS, Hardy SA, Blakely EL, Hopton S, Nemeth AH, Fratter C, Poulton J, Taylor RW, and Downes SM
- Subjects
- Adult, DNA Mutational Analysis, Electron Transport Complex IV metabolism, Electrooculography, Electroretinography, Humans, Male, Mitochondria, Muscle enzymology, Mitochondria, Muscle genetics, Mitochondria, Muscle pathology, Muscle, Skeletal enzymology, Muscle, Skeletal pathology, Optical Imaging, Succinate Dehydrogenase metabolism, Usher Syndromes diagnosis, Usher Syndromes enzymology, DNA, Mitochondrial genetics, Photoreceptor Cells, Vertebrate pathology, Point Mutation, RNA, Transfer, Lys genetics, Thymidine Kinase genetics, Usher Syndromes genetics
- Abstract
Background/aim: The rare mitochondrial DNA (mtDNA) variant m.8340G>A has been previously reported in the literature in a single, sporadic case of mitochondrial myopathy. In this report, we aim to investigate the case of a 39-year-old male patient with sensorineural deafness who presented to the eye clinic with nyctalopia, retinal pigmentary changes and bilateral cortical cataracts., Methods: The patient was examined clinically and investigated with autofluorescence, full-field electroretinography, electro-oculogram and dark adaptometry. Sequencing of the mitochondrial genome in blood and muscle tissue was followed by histochemical and biochemical analyses together with single fibre studies of a muscle biopsy to confirm a mitochondrial aetiology., Results: Electrophysiology, colour testing and dark adaptometry showed significant photoreceptor dysfunction with macular involvement. Sequencing the complete mitochondrial genome revealed a rare mitochondrial tRNA
Lys ( MTTK ) gene variant-m.8340G>A-which was heteroplasmic in blood (11%) and skeletal muscle (65%) and cosegregated with cytochrome c oxidase-deficient fibres in single-fibre studies., Conclusion: We confirm the pathogenicity of the rare mitochondrial m.8340G>A variant the basis of single-fibre segregation studies and its association with an expanded clinical phenotype. Our case expands the phenotypic spectrum of diseases associated with mitochondrial tRNA point mutations, highlighting the importance of considering a mitochondrial diagnosis in similar cases presenting to the eye clinic and the importance of further genetic testing if standard mutational analysis does not yield a result., Competing Interests: Competing interests: RWT is supported by a Wellcome Trust Strategic Award (096919/Z/11/Z), the MRC Centre for Neuromuscular Diseases (G0601943), the Lily Foundation and the UK NHS Highly Specialised “Rare Mitochondrial Disorders of Adults and Children” Service in Newcastle upon Tyne., (© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2017. All rights reserved. No commercial use is permitted unless otherwise expressly granted.)- Published
- 2017
- Full Text
- View/download PDF
229. A novel mutation in the MYO7A gene is associated with Usher syndrome type 1 in a Chinese family.
- Author
-
He X, Peng Q, Li S, Zhu P, Wu C, Rao C, Lin J, and Lu X
- Subjects
- Adult, Asian People genetics, Audiometry, Child, DNA Mutational Analysis, Family, Female, Hearing Loss, Sensorineural, Heterozygote, High-Throughput Nucleotide Sequencing, Humans, Male, Mutation, Myosin VIIa, Pedigree, Phenotype, Myosins genetics, Usher Syndromes genetics
- Abstract
Objectives: We aimed to investigate the genetic causes of hearing loss in a Chinese proband with autosomal recessive congenital deafness., Methods: The targeted capture of 159 known deafness genes and next-generation sequencing were performed to study the genetic causes of hearing loss in the Chinese family. Sanger sequencing was employed to verify the variant mutations in members of this family., Results: The proband harbored two mutations in the MYO7A gene in the form of compound heterozygosity. She was found to be heterozygous for a novel insertion mutation c.3847_3848 ins TCTG (p.N1285LfsX24) in exon 30 and for the known mutation c.2239_2240delAG (p.R747S fsX16)in exon 19. The novel mutation was absent in the 1000 Genomes Project. These variants were carried in the heterozygous state by the parents and were therefore co-segregated with the genetic disease. Clinical re-assessment, including detailed audiologic and ocular examinations, revealed congenital deafness and retinitis pigmentosa in the proband. Collectively, the combination of audiometric, ophthalmologic and genetic examinations successfully confirmed the phenotype of Usher syndrome type 1 (USH1)., Conclusion: This study demonstrates that the novel mutation c.3847_3848insTCTG (p. N1285LfsX24) in compound heterozygosity with c.2239_2240delAG in the MYO7A gene is the main cause of USH1 in the proband. Our study expands the mutational spectrum of MYO7A and provides a foundation for further investigations elucidating the MYO7A-related mechanisms of USH1., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
230. CLINICAL PRESENTATION AND DISEASE COURSE OF USHER SYNDROME BECAUSE OF MUTATIONS IN MYO7A OR USH2A.
- Author
-
Testa F, Melillo P, Bonnet C, Marcelli V, de Benedictis A, Colucci R, Gallo B, Kurtenbach A, Rossi S, Marciano E, Auricchio A, Petit C, Zrenner E, and Simonelli F
- Subjects
- Adolescent, Adult, DNA Mutational Analysis, Disease Progression, Electroretinography, Extracellular Matrix Proteins metabolism, Female, Follow-Up Studies, Humans, Male, Middle Aged, Myosin VIIa, Myosins metabolism, Phenotype, Retina diagnostic imaging, Retina physiopathology, Retrospective Studies, Time Factors, Tomography, Optical Coherence, Usher Syndromes diagnosis, Usher Syndromes physiopathology, Young Adult, DNA genetics, Extracellular Matrix Proteins genetics, Mutation, Myosins genetics, Usher Syndromes genetics, Visual Acuity, Visual Fields
- Abstract
Purpose: To evaluate differences in the visual phenotype and natural history of Usher syndrome caused by mutations in MYO7A or USH2A, the most commonly affected genes of Usher syndrome Type I (USH1) and Type II (USH2), respectively., Methods: Eighty-eight patients with a clinical diagnosis of USH1 (26 patients) or USH2 (62 patients) were retrospectively evaluated. Of these, 48 patients had 2 disease-causing mutations in MYO7A (10 USH1 patients), USH2A (33 USH2 patients), and other USH (5 patients) genes. Clinical investigation included best-corrected visual acuity, Goldmann visual field, fundus photography, electroretinography, and audiologic and vestibular assessments. Longitudinal analysis was performed over a median follow-up time of 3.5 years., Results: Patients carrying mutations in MYO7A had a younger age of onset of hearing and visual impairments than those carrying mutations in USH2A, leading to an earlier diagnosis of the disease in the former patients. Longitudinal analysis showed that visual acuity and visual field decreased more rapidly in subjects carrying MYO7A mutations than in those carrying USH2A mutations (mean annual exponential rates of decline of 3.92 vs. 3.44% and of 8.52 vs. 4.97%, respectively), and the former patients reached legal blindness on average 15 years earlier than the latter., Conclusion: The current study confirmed a more severe progression of the retinal disease in USH1 patients rather than in USH2 patients. Furthermore, most visual symptoms (i.e., night blindness, visual acuity worsening) occurred at an earlier age in USH1 patients carrying mutations in MYO7A.
- Published
- 2017
- Full Text
- View/download PDF
231. The Usher Syndrome Type IIIB Histidyl-tRNA Synthetase Mutation Confers Temperature Sensitivity.
- Author
-
Abbott JA, Guth E, Kim C, Regan C, Siu VM, Rupar CA, Demeler B, Francklyn CS, and Robey-Bond SM
- Subjects
- Amino Acid Sequence, Aminoacylation, Cells, Cultured, Enzyme Stability, HEK293 Cells, Histidine-tRNA Ligase chemistry, Humans, Kinetics, Models, Molecular, Protein Biosynthesis, RNA, Transfer metabolism, Sequence Alignment, Temperature, Usher Syndromes metabolism, Histidine-tRNA Ligase genetics, Histidine-tRNA Ligase metabolism, Point Mutation, Usher Syndromes enzymology, Usher Syndromes genetics
- Abstract
Histidyl-tRNA synthetase (HARS) is a highly conserved translation factor that plays an essential role in protein synthesis. HARS has been implicated in the human syndromes Charcot-Marie-Tooth (CMT) Type 2W and Type IIIB Usher (USH3B). The USH3B mutation, which encodes a Y454S substitution in HARS, is inherited in an autosomal recessive fashion and associated with childhood deafness, blindness, and episodic hallucinations during acute illness. The biochemical basis of the pathophysiologies linked to USH3B is currently unknown. Here, we present a detailed functional comparison of wild-type (WT) and Y454S HARS enzymes. Kinetic parameters for enzymes and canonical substrates were determined using both steady state and rapid kinetics. Enzyme stability was examined using differential scanning fluorimetry. Finally, enzyme functionality in a primary cell culture was assessed. Our results demonstrate that the Y454S substitution leaves HARS amino acid activation, aminoacylation, and tRNA
His binding functions largely intact compared with those of WT HARS, and the mutant enzyme dimerizes like the wild type does. Interestingly, during our investigation, it was revealed that the kinetics of amino acid activation differs from that of the previously characterized bacterial HisRS. Despite the similar kinetics, differential scanning fluorimetry revealed that Y454S is less thermally stable than WT HARS, and cells from Y454S patients grown at elevated temperatures demonstrate diminished levels of protein synthesis compared to those of WT cells. The thermal sensitivity associated with the Y454S mutation represents a biochemical basis for understanding USH3B.- Published
- 2017
- Full Text
- View/download PDF
232. Human myosin VIIa is a very slow processive motor protein on various cellular actin structures.
- Author
-
Sato O, Komatsu S, Sakai T, Tsukasaki Y, Tanaka R, Mizutani T, Watanabe TM, Ikebe R, and Ikebe M
- Subjects
- 3T3 Cells, Actins genetics, Amino Acid Sequence, Animals, Humans, Mice, Myosin Heavy Chains genetics, Myosin Heavy Chains metabolism, Myosin Type V genetics, Myosin Type V metabolism, Myosin VIIa, Myosins genetics, Protein Transport genetics, Pseudopodia genetics, Sequence Deletion, Usher Syndromes genetics, Actins metabolism, Myosins metabolism, Pseudopodia metabolism, Usher Syndromes metabolism
- Abstract
Human myosin VIIa (MYO7A) is an actin-linked motor protein associated with human Usher syndrome (USH) type 1B, which causes human congenital hearing and visual loss. Although it has been thought that the role of human myosin VIIa is critical for USH1 protein tethering with actin and transportation along actin bundles in inner-ear hair cells, myosin VIIa's motor function remains unclear. Here, we studied the motor function of the tail-truncated human myosin VIIa dimer (HM7AΔTail/LZ) at the single-molecule level. We found that the HM7AΔTail/LZ moves processively on single actin filaments with a step size of 35 nm. Dwell-time distribution analysis indicated an average waiting time of 3.4 s, yielding ∼0.3 s
-1 for the mechanical turnover rate; hence, the velocity of HM7AΔTail/LZ was extremely slow, at 11 nm·s-1 We also examined HM7AΔTail/LZ movement on various actin structures in demembranated cells. HM7AΔTail/LZ showed unidirectional movement on actin structures at cell edges, such as lamellipodia and filopodia. However, HM7AΔTail/LZ frequently missed steps on actin tracks and exhibited bidirectional movement at stress fibers, which was not observed with tail-truncated myosin Va. These results suggest that the movement of the human myosin VIIa motor protein is more efficient on lamellipodial and filopodial actin tracks than on stress fibers, which are composed of actin filaments with different polarity, and that the actin structures influence the characteristics of cargo transportation by human myosin VIIa. In conclusion, myosin VIIa movement appears to be suitable for translocating USH1 proteins on stereocilia actin bundles in inner-ear hair cells., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2017
- Full Text
- View/download PDF
233. Usher syndrome type 1-associated cadherins shape the photoreceptor outer segment.
- Author
-
Schietroma C, Parain K, Estivalet A, Aghaie A, Boutet de Monvel J, Picaud S, Sahel JA, Perron M, El-Amraoui A, and Petit C
- Subjects
- Actin Cytoskeleton metabolism, Animals, Cadherins genetics, Gene Expression Regulation, Developmental, Gene Knockdown Techniques, Larva genetics, Larva metabolism, Retinal Cone Photoreceptor Cells ultrastructure, Retinal Photoreceptor Cell Outer Segment ultrastructure, Rod Cell Outer Segment ultrastructure, Usher Syndromes genetics, Usher Syndromes pathology, Xenopus embryology, Xenopus genetics, Xenopus Proteins genetics, Cadherins metabolism, Retinal Cone Photoreceptor Cells metabolism, Retinal Photoreceptor Cell Outer Segment metabolism, Rod Cell Outer Segment metabolism, Usher Syndromes metabolism, Xenopus metabolism, Xenopus Proteins metabolism
- Abstract
Usher syndrome type 1 (USH1) causes combined hearing and sight defects, but how mutations in USH1 genes lead to retinal dystrophy in patients remains elusive. The USH1 protein complex is associated with calyceal processes, which are microvilli of unknown function surrounding the base of the photoreceptor outer segment. We show that in Xenopus tropicalis , these processes are connected to the outer-segment membrane by links composed of protocadherin-15 (USH1F protein). Protocadherin-15 deficiency, obtained by a knockdown approach, leads to impaired photoreceptor function and abnormally shaped photoreceptor outer segments. Rod basal outer disks displayed excessive outgrowth, and cone outer segments were curved, with lamellae of heterogeneous sizes, defects also observed upon knockdown of Cdh23 , encoding cadherin-23 (USH1D protein). The calyceal processes were virtually absent in cones and displayed markedly reduced F-actin content in rods, suggesting that protocadherin-15-containing links are essential for their development and/or maintenance. We propose that calyceal processes, together with their associated links, control the sizing of rod disks and cone lamellae throughout their daily renewal., (© 2017 Schietroma et al.)
- Published
- 2017
- Full Text
- View/download PDF
234. Novel compound heterozygous MYO7A mutations in Moroccan families with autosomal recessive non-syndromic hearing loss.
- Author
-
Bakhchane A, Charif M, Bousfiha A, Boulouiz R, Nahili H, Rouba H, Charoute H, Lenaers G, and Barakat A
- Subjects
- Adult, Exome, Female, Humans, Male, Models, Molecular, Morocco, Myosin VIIa, Myosins chemistry, Pedigree, Heterozygote, Mutation, Myosins genetics, Usher Syndromes genetics
- Abstract
The MYO7A gene encodes a protein belonging to the unconventional myosin super family. Mutations within MYO7A can lead to either non syndromic hearing loss or to the Usher syndrome type 1B (USH1B). Here, we report the results of genetic analyses performed on Moroccan families with autosomal recessive non syndromic hearing loss that identified two families with compound heterozygous MYO7A mutations. Five mutations (c.6025delG, c.6229T>A, c.3500T>A, c.5617C>T and c.4487C>A) were identified in these families, the latter presenting two differently affected branches. Multiple bioinformatics programs and molecular modelling predicted the pathogenic effect of these mutations. In conclusion, the absence of vestibular and retinal symptom in the affected patients suggests that these families have the isolated non-syndromic hearing loss DFNB2 (nonsyndromic autosomal recessive hearing loss) presentation, instead of USH1B.
- Published
- 2017
- Full Text
- View/download PDF
235. A deep intronic CLRN1 (USH3A) founder mutation generates an aberrant exon and underlies severe Usher syndrome on the Arabian Peninsula.
- Author
-
Khan AO, Becirovic E, Betz C, Neuhaus C, Altmüller J, Maria Riedmayr L, Motameny S, Nürnberg G, Nürnberg P, and Bolz HJ
- Subjects
- Consanguinity, DNA Mutational Analysis, Exons, Genetic Linkage, Genetic Predisposition to Disease, Genome-Wide Association Study, Genotype, Humans, Mutation, Pedigree, Saudi Arabia, Membrane Proteins genetics, Usher Syndromes genetics
- Abstract
Deafblindness is mostly due to Usher syndrome caused by recessive mutations in the known genes. Mutation-negative patients therefore either have distinct diseases, mutations in yet unknown Usher genes or in extra-exonic parts of the known genes - to date a largely unexplored possibility. In a consanguineous Saudi family segregating Usher syndrome type 1 (USH1), NGS of genes for Usher syndrome, deafness and retinal dystrophy and subsequent whole-exome sequencing each failed to identify a mutation. Genome-wide linkage analysis revealed two small candidate regions on chromosome 3, one containing the USH3A gene CLRN1, which has never been associated with Usher syndrome in Saudi Arabia. Whole-genome sequencing (WGS) identified a homozygous deep intronic mutation, c.254-649T > G, predicted to generate a novel donor splice site. CLRN1 minigene-based analysis confirmed the splicing of an aberrant exon due to usage of this novel motif, resulting in a frameshift and a premature termination codon. We identified this mutation in an additional two of seven unrelated mutation-negative Saudi USH1 patients. Locus-specific markers indicated that c.254-649T > G
CLRN1 represents a founder allele that may significantly contribute to deafblindness in this population. Our finding underlines the potential of WGS to uncover atypically localized, hidden mutations in patients who lack exonic mutations in the known disease genes.- Published
- 2017
- Full Text
- View/download PDF
236. Laser-capture micro dissection combined with next-generation sequencing analysis of cell type-specific deafness gene expression in the mouse cochlea.
- Author
-
Nishio SY, Takumi Y, and Usami SI
- Subjects
- Animals, Cochlear Implantation adverse effects, Deafness diagnosis, Disease Models, Animal, Gene Expression Profiling, High-Throughput Nucleotide Sequencing, Immunohistochemistry, In Situ Hybridization, Male, Mice, Mice, Inbred C57BL, Oligonucleotide Array Sequence Analysis, Polymerase Chain Reaction, Usher Syndromes genetics, Cochlea metabolism, Deafness genetics, Laser Capture Microdissection, Spiral Ganglion pathology
- Abstract
Cochlear implantation (CI), which directly stimulates the cochlear nerves, is the most effective and widely used medical intervention for patients with severe to profound sensorineural hearing loss. The etiology of the hearing loss is speculated to have a major influence of CI outcomes, particularly in cases resulting from mutations in genes preferentially expressed in the spiral ganglion region. To elucidate precise gene expression levels in each part of the cochlea, we performed laser-capture micro dissection in combination with next-generation sequencing analysis and determined the expression levels of all known deafness-associated genes in the organ of Corti, spiral ganglion, lateral wall, and spiral limbs. The results were generally consistent with previous reports based on immunocytochemistry or in situ hybridization. As a notable result, the genes associated with many kinds of syndromic hearing loss (such as Clpp, Hars2, Hsd17b4, Lars2 for Perrault syndrome, Polr1c and Polr1d for Treacher Collins syndrome, Ndp for Norrie Disease, Kal for Kallmann syndrome, Edn3 and Snai2 for Waardenburg Syndrome, Col4a3 for Alport syndrome, Sema3e for CHARGE syndrome, Col9a1 for Sticker syndrome, Cdh23, Cib2, Clrn1, Pcdh15, Ush1c, Ush2a, Whrn for Usher syndrome and Wfs1 for Wolfram syndrome) showed higher levels of expression in the spiral ganglion than in other parts of the cochlea. This dataset will provide a base for more detailed analysis in order to clarify gene functions in the cochlea as well as predict CI outcomes based on gene expression data., (Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
237. Advances in genetic hearing loss: CIB2 gene.
- Author
-
Jacoszek A, Pollak A, Płoski R, and Ołdak M
- Subjects
- Animals, Calcium metabolism, Calcium-Binding Proteins metabolism, Ear, Inner pathology, Hearing Loss, Sensorineural genetics, Humans, Signal Transduction, Usher Syndromes genetics, Calcium-Binding Proteins genetics, Hearing Loss genetics, Mutation
- Abstract
Hearing plays a crucial role in human development. Receiving and processing sounds are essential for the advancement of the speech ability during the early childhood and for a proper functioning in the society. Hearing loss is one of the most frequent disabilities that affect human senses. It can be caused by genetic or environmental factors or both of them. Calcium- and integrin-binding protein 2 (CIB2) is one of the recently identified genes, involved in HI pathogenesis. CIB2 is widely expressed in various human and animal tissues, mainly in skeletal muscle, nervous tissue, inner ear, and retina. The CIB2 protein is responsible for maintaining Ca
2+ homeostasis in cells and interacting with integrins-transmembrane receptors essential for cell adhesion, migration, and activation of signaling pathways. Calcium signaling pathway is crucial for signal transduction in the inner ear, and integrins regulate hair cell differentiation and maturation of the stereocilia. To date, mutations detected in CIB2 are causative for nonsyndromic hearing loss (DFNB48) or Usher syndrome type 1 J. Patients harboring biallelic CIB2 mutations suffer from bilateral, early onset, moderate to profound HI. In the paper, we summarize the current status of the research on CIB2.- Published
- 2017
- Full Text
- View/download PDF
238. Genetic analysis of 10 pedigrees with inherited retinal degeneration by exome sequencing and phenotype-genotype association.
- Author
-
Biswas P, Duncan JL, Maranhao B, Kozak I, Branham K, Gabriel L, Lin JH, Barteselli G, Navani M, Suk J, Parke M, Schlechter C, Weleber RG, Heckenlively JR, Dagnelie G, Lee P, Riazuddin SA, and Ayyagari R
- Subjects
- ADP-Ribosylation Factors genetics, ATP-Binding Cassette Transporters genetics, Alkyl and Aryl Transferases genetics, DNA Mutational Analysis, Female, Genetic Association Studies, Humans, Male, Mutation genetics, Pedigree, Phosphotransferases (Alcohol Group Acceptor) genetics, Usher Syndromes genetics, cis-trans-Isomerases genetics, Exome genetics, Genotype, Phenotype, Retinal Degeneration genetics
- Abstract
Our purpose was to identify causative mutations and characterize the phenotype associated with the genotype in 10 unrelated families with autosomal recessive retinal degeneration. Ophthalmic evaluation and DNA isolation were carried out in 10 pedigrees with inherited retinal degenerations (IRD). Exomes of probands from eight pedigrees were captured using Nimblegen V2/V3 or Agilent V5+UTR kits, and sequencing was performed on Illumina HiSeq. The DHDDS gene was screened for mutations in the remaining two pedigrees with Ashkenazi Jewish ancestry. Exome variants were filtered to detect candidate causal variants using exomeSuite software. Segregation and ethnicity-matched control sample analysis were performed by dideoxy sequencing. Retinal histology of a patient with DHDDS mutation was studied by microscopy. Genetic analysis identified six known mutations in ABCA4 (p.Gly1961Glu, p.Ala1773Val, c.5461-10T>C), RPE65 (p.Tyr249Cys, p.Gly484Asp), PDE6B (p.Lys706Ter) and DHDDS (p.Lys42Glu) and ten novel potentially pathogenic variants in CERKL (p.Met323Val fsX20), RPE65 (p.Phe252Ser, Thr454Leu fsX31), ARL6 (p.Arg121His), USH2A (p.Gly3142Ter, p.Cys3294Trp), PDE6B (p.Gln652Ter), and DHDDS (p.Thr206Ala) genes. Among these, variants/mutations in two separate genes were observed to segregate with IRD in two pedigrees. Retinal histopathology of a patient with a DHDDS mutation showed severe degeneration of retinal layers with relative preservation of the retinal pigment epithelium. Analysis of exome variants in ten pedigrees revealed nine novel potential disease-causing variants and nine previously reported homozygous or compound heterozygous mutations in the CERKL , ABCA4, RPE65 , ARL6, USH2A , PDE6B , and DHDDS genes. Mutations that could be sufficient to cause pathology were observed in more than one gene in one pedigree., (Copyright © 2017 the American Physiological Society.)
- Published
- 2017
- Full Text
- View/download PDF
239. Characterization of the ternary Usher syndrome SANS/ush2a/whirlin protein complex.
- Author
-
Sorusch N, Bauß K, Plutniok J, Samanta A, Knapp B, Nagel-Wolfrum K, and Wolfrum U
- Subjects
- Deaf-Blind Disorders pathology, Extracellular Matrix Proteins chemistry, Extracellular Matrix Proteins metabolism, Hair Cells, Auditory metabolism, Hair Cells, Auditory pathology, Humans, Membrane Proteins chemistry, Multiprotein Complexes chemistry, Multiprotein Complexes genetics, Multiprotein Complexes metabolism, Mutation, Nerve Tissue Proteins chemistry, Nerve Tissue Proteins metabolism, Photoreceptor Cells metabolism, Photoreceptor Cells pathology, Protein Binding, Protein Interaction Maps genetics, Protein Structure, Tertiary, Usher Syndromes complications, Usher Syndromes pathology, Deaf-Blind Disorders genetics, Extracellular Matrix Proteins genetics, Membrane Proteins genetics, Nerve Tissue Proteins genetics, Usher Syndromes genetics
- Abstract
The Usher syndrome (USH) is the most common form of inherited deaf-blindness, accompanied by vestibular dysfunction. Due to the heterogeneous manifestation of the clinical symptoms, three USH types (USH1-3) and additional atypical forms are distinguished. USH1 and USH2 proteins have been shown to function together in multiprotein networks in photoreceptor cells and hair cells. Mutations in USH proteins are considered to disrupt distinct USH protein networks and finally lead to the development of USH.To get novel insights into the molecular pathomechanisms underlying USH, we further characterize the periciliary USH protein network in photoreceptor cells. We show the direct interaction between the scaffold protein SANS (USH1G) and the transmembrane adhesion protein ush2a and that both assemble into a ternary USH1/USH2 complex together with the PDZ-domain protein whirlin (USH2D) via mutual interactions. Immunohistochemistry and proximity ligation assays demonstrate co-localization of complex partners and complex formation, respectively, in the periciliary region, the inner segment and at the synapses of rodent and human photoreceptor cells. Protein-protein interaction assays and co-expression of complex partners reveal that pathogenic mutations in USH1G severely affect formation of the SANS/ush2a/whirlin complex. Translational read-through drug treatment, targeting the c.728C > A (p.S243X) nonsense mutation, restored SANS scaffold function. We conclude that USH1 and USH2 proteins function together in higher order protein complexes. The maintenance of USH1/USH2 protein complexes depends on multiple USH1/USH2 protein interactions, which are disrupted by pathogenic mutations in USH1G protein SANS., (© The Author 2017. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
240. Gene therapy restores auditory and vestibular function in a mouse model of Usher syndrome type 1c.
- Author
-
Pan B, Askew C, Galvin A, Heman-Ackah S, Asai Y, Indzhykulian AA, Jodelka FM, Hastings ML, Lentz JJ, Vandenberghe LH, Holt JR, and Géléoc GS
- Subjects
- Animals, Cell Cycle Proteins, Cytoskeletal Proteins, Female, Gene Knock-In Techniques, Hearing Loss, Sensorineural diagnosis, Hearing Loss, Sensorineural genetics, Male, Mice, Mice, Inbred C57BL, Plasmids administration & dosage, Plasmids genetics, Recovery of Function genetics, Treatment Outcome, Vestibular Diseases diagnosis, Vestibular Diseases genetics, Carrier Proteins genetics, Genetic Therapy methods, Hearing Loss, Sensorineural therapy, Usher Syndromes genetics, Usher Syndromes therapy, Vestibular Diseases therapy
- Abstract
Because there are currently no biological treatments for hearing loss, we sought to advance gene therapy approaches to treat genetic deafness. We focused on Usher syndrome, a devastating genetic disorder that causes blindness, balance disorders and profound deafness, and studied a knock-in mouse model, Ush1c c.216G>A, for Usher syndrome type IC (USH1C). As restoration of complex auditory and balance function is likely to require gene delivery systems that target auditory and vestibular sensory cells with high efficiency, we delivered wild-type Ush1c into the inner ear of Ush1c c.216G>A mice using a synthetic adeno-associated viral vector, Anc80L65, shown to transduce 80-90% of sensory hair cells. We demonstrate recovery of gene and protein expression, restoration of sensory cell function, rescue of complex auditory function and recovery of hearing and balance behavior to near wild-type levels. The data represent unprecedented recovery of inner ear function and suggest that biological therapies to treat deafness may be suitable for translation to humans with genetic inner ear disorders.
- Published
- 2017
- Full Text
- View/download PDF
241. Gene Therapy Restores Balance and Auditory Functions in a Mouse Model of Usher Syndrome.
- Author
-
Isgrig K, Shteamer JW, Belyantseva IA, Drummond MC, Fitzgerald TS, Vijayakumar S, Jones SM, Griffith AJ, Friedman TB, Cunningham LL, and Chien WW
- Subjects
- Animals, Behavior, Animal, Disease Models, Animal, Gene Expression, Hair Cells, Auditory, Inner metabolism, Hair Cells, Auditory, Inner ultrastructure, Hearing Tests, Humans, Membrane Proteins genetics, Mice, Mice, Knockout, Phenotype, Stereocilia metabolism, Stereocilia ultrastructure, Usher Syndromes therapy, Genetic Therapy, Hearing genetics, Postural Balance genetics, Usher Syndromes genetics, Usher Syndromes physiopathology
- Abstract
Dizziness and hearing loss are among the most common disabilities. Many forms of hereditary balance and hearing disorders are caused by abnormal development of stereocilia, mechanosensory organelles on the apical surface of hair cells in the inner ear. The deaf whirler mouse, a model of human Usher syndrome (manifested by hearing loss, dizziness, and blindness), has a recessive mutation in the whirlin gene, which renders hair cell stereocilia short and dysfunctional. In this study, wild-type whirlin cDNA was delivered to the inner ears of neonatal whirler mice using adeno-associated virus serotype 2/8 (AAV8-whirlin) by injection into the posterior semicircular canal. Unilateral whirlin gene therapy injection was able to restore balance function as well as improve hearing in whirler mice for at least 4 months. Our data indicate that gene therapy is likely to become a treatment option for hereditary disorders of balance and hearing., (Copyright © 2017. Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
242. CEP78 is mutated in a distinct type of Usher syndrome.
- Author
-
Fu Q, Xu M, Chen X, Sheng X, Yuan Z, Liu Y, Li H, Sun Z, Li H, Yang L, Wang K, Zhang F, Li Y, Zhao C, Sui R, and Chen R
- Subjects
- Adult, Child, Consanguinity, Exome genetics, Female, Frameshift Mutation, Genome, Human, Hair Cells, Auditory, Inner pathology, Homozygote, Humans, Male, Pedigree, Retinitis Pigmentosa pathology, Usher Syndromes pathology, Cell Cycle Proteins genetics, High-Throughput Nucleotide Sequencing, Retinitis Pigmentosa genetics, Usher Syndromes genetics
- Abstract
Background: Usher syndrome is a genetically heterogeneous disorder featured by combined visual impairment and hearing loss. Despite a dozen of genes involved in Usher syndrome having been identified, the genetic basis remains unknown in 20-30% of patients. In this study, we aimed to identify the novel disease-causing gene of a distinct subtype of Usher syndrome., Methods: Ophthalmic examinations and hearing tests were performed on patients with Usher syndrome in two consanguineous families. Target capture sequencing was initially performed to screen causative mutations in known retinal disease-causing loci. Whole exome sequencing (WES) and whole genome sequencing (WGS) were applied for identifying novel disease-causing genes. RT-PCR and Sanger sequencing were performed to evaluate the splicing-altering effect of identified CEP78 variants., Results: Patients from the two independent families show a mild Usher syndrome phenotype featured by juvenile or adult-onset cone-rod dystrophy and sensorineural hearing loss. WES and WGS identified two homozygous rare variants that affect mRNA splicing of a ciliary gene CEP78 . RT-PCR confirmed that the two variants indeed lead to abnormal splicing, resulting in premature stop of protein translation due to frameshift., Conclusions: Our results provide evidence that CEP78 is a novel disease-causing gene for Usher syndrome, demonstrating an additional link between ciliopathy and Usher protein network in photoreceptor cells and inner ear hair cells., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)
- Published
- 2017
- Full Text
- View/download PDF
243. Gene therapy approaches for prevention of retinal degeneration in Usher syndrome.
- Author
-
Williams DS, Chadha A, Hazim R, and Gibbs D
- Subjects
- Humans, Retinal Degeneration genetics, Usher Syndromes therapy, Genetic Therapy methods, Retinal Degeneration therapy, Usher Syndromes genetics
- Published
- 2017
- Full Text
- View/download PDF
244. The roles of USH1 proteins and PDZ domain-containing USH proteins in USH2 complex integrity in cochlear hair cells.
- Author
-
Zou J, Chen Q, Almishaal A, Mathur PD, Zheng T, Tian C, Zheng QY, and Yang J
- Subjects
- Animals, Carrier Proteins chemistry, Cell Cycle Proteins, Cytoskeletal Proteins, Extracellular Matrix Proteins chemistry, Hair Cells, Auditory pathology, Humans, Mice, Multiprotein Complexes chemistry, Multiprotein Complexes genetics, Myosin VIIa, Myosins chemistry, PDZ Domains genetics, Retina metabolism, Retina pathology, Stereocilia genetics, Stereocilia metabolism, Stereocilia pathology, Usher Syndromes pathology, Carrier Proteins genetics, Extracellular Matrix Proteins genetics, Myosins genetics, Usher Syndromes genetics
- Abstract
Usher syndrome (USH) is the most common cause of inherited deaf-blindness, manifested as USH1, USH2 and USH3 clinical types. The protein products of USH2 causative and modifier genes, USH2A, ADGRV1, WHRN and PDZD7, interact to assemble a multiprotein complex at the ankle link region of the mechanosensitive stereociliary bundle in hair cells. Defects in this complex cause stereociliary bundle disorganization and hearing loss. The four USH2 proteins also interact in vitro with USH1 proteins including myosin VIIa, USH1G (SANS), CIB2 and harmonin. However, it is unclear whether the interactions between USH1 and USH2 proteins occur in vivo and whether USH1 proteins play a role in USH2 complex assembly in hair cells. In this study, we identified a novel interaction between myosin VIIa and PDZD7 by FLAG pull-down assay. We further investigated the role of the above-mentioned four USH1 proteins in the cochlear USH2 complex assembly using USH1 mutant mice. We showed that only myosin VIIa is indispensable for USH2 complex assembly at ankle links, indicating the potential transport and/or anchoring role of myosin VIIa for USH2 proteins in hair cells. However, myosin VIIa is not required for USH2 complex assembly in photoreceptors. We further showed that, while PDZ protein harmonin is not involved, its paralogous USH2 proteins, PDZD7 and whirlin, function synergistically in USH2 complex assembly in cochlear hair cells. In summary, our studies provide novel insight into the functional relationship between USH1 and USH2 proteins in the cochlea and the retina as well as the disease mechanisms underlying USH1 and USH2., (© The Author 2016. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
245. Adenocarcinoma of the Rectum in a 27-year-old Patient with Usher Syndrome : Is there a Genetic Correlation?
- Author
-
Helsloot L, Reynders D, Cools P, and Wauters J
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adenocarcinoma complications, Adult, Cell Cycle Proteins, Cytoskeletal Proteins, Female, Humans, Proteins genetics, Rectal Neoplasms complications, Usher Syndromes complications, Adenocarcinoma genetics, Rectal Neoplasms genetics, Usher Syndromes genetics
- Published
- 2017
246. An innovative strategy for the molecular diagnosis of Usher syndrome identifies causal biallelic mutations in 93% of European patients.
- Author
-
Bonnet C, Riahi Z, Chantot-Bastaraud S, Smagghe L, Letexier M, Marcaillou C, Lefèvre GM, Hardelin JP, El-Amraoui A, Singh-Estivalet A, Mohand-Saïd S, Kohl S, Kurtenbach A, Sliesoraityte I, Zobor D, Gherbi S, Testa F, Simonelli F, Banfi S, Fakin A, Glavač D, Jarc-Vidmar M, Zupan A, Battelino S, Martorell Sampol L, Claveria MA, Catala Mora J, Dad S, Møller LB, Rodriguez Jorge J, Hawlina M, Auricchio A, Sahel JA, Marlin S, Zrenner E, Audo I, and Petit C
- Subjects
- Alleles, Comparative Genomic Hybridization methods, Europe, Exome, Extracellular Matrix Proteins genetics, Genes, Modifier, Humans, Sensitivity and Specificity, Sequence Analysis, DNA methods, Usher Syndromes diagnosis, Genetic Testing methods, Mutation, Usher Syndromes genetics
- Abstract
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. We screened a large cohort of 427 patients (139 USH1, 282 USH2, and six of undefined clinical subtype) from various European medical centers for mutations in all USH genes and the modifier gene. We identified a total of 421 different sequence variants predicted to be pathogenic, about half of which had not been previously reported. Remarkably, we detected large genomic rearrangements, most of which were novel and unique, in 9% of the patients. Thus, our strategy led to the identification of biallelic and monoallelic mutations in 92.7% and 5.8% of the USH patients, respectively. With an overall 98.5% mutation characterization rate, the diagnosis efficiency was substantially improved compared with previously reported methods.
- Published
- 2016
- Full Text
- View/download PDF
247. Compound heterozygous MYO7A mutations segregating Usher syndrome type 2 in a Han family.
- Author
-
Zong L, Chen K, Wu X, Liu M, and Jiang H
- Subjects
- Adolescent, Adult, Child, DNA Mutational Analysis, Female, Heterozygote, High-Throughput Nucleotide Sequencing, Humans, Male, Middle Aged, Mutation, Myosin VIIa, Pedigree, Phenotype, Young Adult, Asian People genetics, Family, Myosins genetics, Usher Syndromes genetics
- Abstract
Objective: Identification of rare deafness genes for inherited congenital sensorineural hearing impairment remains difficult, because a large variety of genes are implicated. In this study we applied targeted capture and next-generation sequencing to uncover the underlying gene in a three-generation Han family segregating recessive inherited hearing loss and retinitis pigmentosa., Methods: After excluding mutations in common deafness genes GJB2, SLC26A4 and the mitochondrial gene, genomic DNA of the proband of a Han family was subjected to targeted next-generation sequencing. The candidate mutations were confirmed by Sanger sequencing and subsequently analyzed with in silico tools., Results: An unreported splice site mutation c.3924+1G > C compound with c.6028G > A in the MYO7A gene were detected to cosegregate with the phenotype in this pedigree. Both mutations, located in the evolutionarily conserved FERM domain in myosin VIIA, were predicted to be pathogenic. In this family, profound sensorineural hearing impairment and retinitis pigmentosa without vestibular disorder, constituted the typical Usher syndrome type 2., Conclusion: Identification of novel mutation in compound heterozygosity in MYO7A gene revealed the genetic origin of Usher syndrome type 2 in this Han family., (Copyright © 2016 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
248. A combination of two truncating mutations in USH2A causes more severe and progressive hearing impairment in Usher syndrome type IIa.
- Author
-
Hartel BP, Löfgren M, Huygen PL, Guchelaar I, Lo-A-Njoe Kort N, Sadeghi AM, van Wijk E, Tranebjærg L, Kremer H, Kimberling WJ, Cremers CW, Möller C, and Pennings RJ
- Subjects
- Adolescent, Adult, Aged, Audiometry, Audiometry, Pure-Tone, Auditory Threshold, Cross-Sectional Studies, Female, Genetic Association Studies, Genotype, Hearing, Humans, Linear Models, Male, Middle Aged, Netherlands, Phenotype, Retrospective Studies, Sweden, Young Adult, Extracellular Matrix Proteins genetics, Mutation, Usher Syndromes genetics, Usher Syndromes physiopathology
- Abstract
Objectives: Usher syndrome is an inherited disorder that is characterized by hearing impairment (HI), retinitis pigmentosa, and in some cases vestibular dysfunction. Usher syndrome type IIa is caused by mutations in USH2A. HI in these patients is highly heterogeneous and the present study evaluates the effects of different types of USH2A mutations on the audiometric phenotype. Data from two large centres of expertise on Usher Syndrome in the Netherlands and Sweden were combined in order to create a large combined sample of patients to identify possible genotype-phenotype correlations., Design: A retrospective study on HI in 110 patients (65 Dutch and 45 Swedish) genetically diagnosed with Usher syndrome type IIa. We used methods especially designed for characterizing and testing differences in audiological phenotype between patient subgroups. These methods included Age Related Typical Audiograms (ARTA) and a method to evaluate the difference in the degree of HI developed throughout life between subgroups., Results: Cross-sectional linear regression analysis of last-visit audiograms for the best hearing ear demonstrated a gradual decline of hearing over decades. The congenital level of HI was in the range of 16-33 dB at 0.25-0.5 kHz, and in the range of 51-60 dB at 1-8 kHz. The annual threshold deterioration was in the range of 0.4-0.5 dB/year at 0.25-2 kHz and in the range of 0.7-0.8 dB/year at 4-8 kHz. Patients with two truncating mutations, including homozygotes for the common c.2299delG mutation, developed significantly more severe HI throughout life than patients with one truncating mutation combined with one nontruncating mutation, and patients with two nontruncating mutations., Conclusions: The results have direct implications for patient counselling in terms of prognosis of hearing and may serve as baseline measures for future (genetic) therapeutic interventions., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
249. Diversity of the Genes Implicated in Algerian Patients Affected by Usher Syndrome.
- Author
-
Abdi S, Bahloul A, Behlouli A, Hardelin JP, Makrelouf M, Boudjelida K, Louha M, Cheknene A, Belouni R, Rous Y, Merad Z, Selmane D, Hasbelaoui M, Bonnet C, Zenati A, and Petit C
- Subjects
- Algeria, Humans, Mutation, Missense, Genetic Predisposition to Disease, Genetic Variation, Usher Syndromes genetics
- Abstract
Usher syndrome (USH) is an autosomal recessive disorder characterized by a dual sensory impairment affecting hearing and vision. USH is clinically and genetically heterogeneous. Ten different causal genes have been reported. We studied the molecular bases of the disease in 18 unrelated Algerian patients by targeted-exome sequencing, and identified the causal biallelic mutations in all of them: 16 patients carried the mutations at the homozygous state and 2 at the compound heterozygous state. Nine of the 17 different mutations detected in MYO7A (1 of 5 mutations), CDH23 (4 of 7 mutations), PCDH15 (1 mutation), USH1C (1 mutation), USH1G (1 mutation), and USH2A (1 of 2 mutations), had not been previously reported. The deleterious consequences of a missense mutation of CDH23 (p.Asp1501Asn) and the in-frame single codon deletion in USH1G (p.Ala397del) on the corresponding proteins were predicted from the solved 3D-structures of extracellular cadherin (EC) domains of cadherin-23 and the sterile alpha motif (SAM) domain of USH1G/sans, respectively. In addition, we were able to show that the USH1G mutation is likely to affect the binding interface between the SAM domain and USH1C/harmonin. This should spur the use of 3D-structures, not only of isolated protein domains, but also of protein-protein interaction interfaces, to predict the functional impact of mutations detected in the USH genes., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
250. A novel homozygous MYO7A mutation involved in a Venezuelan population with high frequency of USHER1B.
- Author
-
Guzmán HO, Palacios AM, De Almada MI, and Utrera RA
- Subjects
- Adult, Aged, Child, Exons genetics, Female, Homozygote, Humans, Male, Middle Aged, Myosin VIIa, Pedigree, Polymerase Chain Reaction, Sequence Analysis, DNA, Usher Syndromes diagnosis, Usher Syndromes epidemiology, Venezuela epidemiology, Young Adult, Mutation, Myosins genetics, Usher Syndromes genetics
- Abstract
Background: Macanao's population in Venezuela has perhaps the greatest incidence of USH1B known in Latin America (79 cases per 100,000 population); however, until now no mutation in the MYO7A gene had been reported for this population., Materials and Methods: This study aimed to evaluate the entire coding region of the MYO7A gene by direct sequencing of PCR products obtained from patients clinically diagnosed with USH1B., Results: A novel mutation named c.6079_6081del was detected on exon 45 of the MYO7A gene, causing the loss of a single histidine amino acid at codon 2027 (p.H2027del) located within the second FERM domain of the human protein myosin VIIA. Three patients with clinical diagnosis of USH1B were detected positive in homozygosis for the c.6079_6081del mutation; whereas six people from the same affected family were heterozygotes and three other family members were negative., Conclusion: We suggest that this new mutation named c.6079_6081del (p.H2027del) is the main cause of deaf-blindness found in this family clinically diagnosed as USH1B. Additional studies should be performed on this population to determine whether the c.6079_6081del mutation is the main cause of USH1B for the rest of the population.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.