Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Genet Med ; 21(4): 948-954, 2019 04.
Article in English | MEDLINE | ID: mdl-30245514

ABSTRACT

PURPOSE: The aim of this study was to determine the genetic cause of autosomal dominant nonsyndromic hearing loss segregating in a multigenerational family. METHODS: Clinical examination, genome-wide linkage analysis, and exome sequencing were carried out on the family. RESULTS: Affected individuals presented with early-onset progressive mild hearing impairment with a fairly flat, gently downsloping or U-shaped audiogram configuration. Detailed clinical examination excluded any additional symptoms. Linkage analysis detected an interval on chromosome 1p21 with a logarithm of the odds (LOD) score of 8.29: designated locus DFNA37. Exome sequencing identified a novel canonical acceptor splice-site variant c.652-2A>C in the COL11A1 gene within the DFNA37 locus. Genotyping of all 48 family members confirmed segregation of this variant with the deafness phenotype in the extended family. The c.652-2A>C variant is novel, highly conserved, and confirmed in vitro to alter RNA splicing. CONCLUSION: We have identified COL11A1 as the gene responsible for deafness at the DFNA37 locus. Previously, COL11A1 was solely associated with Marshall and Stickler syndromes. This study expands its phenotypic spectrum to include nonsyndromic deafness. The implications of this discovery are valuable in the clinical diagnosis, prognosis, and treatment of patients with COL11A1 pathogenic variants.


Subject(s)
Collagen Type XI/genetics , Deafness/genetics , Genetic Linkage , Protein Isoforms/genetics , Adolescent , Adult , Child , Child, Preschool , Deafness/physiopathology , Exome/genetics , Female , Genotype , Humans , Infant , Infant, Newborn , Male , Pedigree , Phenotype , Exome Sequencing , Young Adult
2.
Hear Res ; 339: 60-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27318125

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.


Subject(s)
Extracellular Matrix Proteins/genetics , Mutation , Usher Syndromes/genetics , Usher Syndromes/physiopathology , 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
3.
PLoS Genet ; 11(3): e1005137, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25816005

ABSTRACT

Hereditary hearing loss is a clinically and genetically heterogeneous disorder. More than 80 genes have been implicated to date, and with the advent of targeted genomic enrichment and massively parallel sequencing (TGE+MPS) the rate of novel deafness-gene identification has accelerated. Here we report a family segregating post-lingual progressive autosomal dominant non-syndromic hearing loss (ADNSHL). After first excluding plausible variants in known deafness-causing genes using TGE+MPS, we completed whole exome sequencing in three hearing-impaired family members. Only a single variant, p.Arg185Pro in HOMER2, segregated with the hearing-loss phenotype in the extended family. This amino acid change alters a highly conserved residue in the coiled-coil domain of HOMER2 that is essential for protein multimerization and the HOMER2-CDC42 interaction. As a scaffolding protein, HOMER2 is involved in intracellular calcium homeostasis and cytoskeletal organization. Consistent with this function, we found robust expression in stereocilia of hair cells in the murine inner ear and observed that over-expression of mutant p.Pro185 HOMER2 mRNA causes anatomical changes of the inner ear and neuromasts in zebrafish embryos. Furthermore, mouse mutants homozygous for the targeted deletion of Homer2 present with early-onset rapidly progressive hearing loss. These data provide compelling evidence that HOMER2 is required for normal hearing and that its sequence alteration in humans leads to ADNSHL through a dominant-negative mode of action.


Subject(s)
Carrier Proteins/genetics , Ear, Inner/metabolism , Exome/genetics , Hearing Loss, Sensorineural/genetics , Animals , Carrier Proteins/biosynthesis , Cochlea/metabolism , Cochlea/pathology , Ear, Inner/pathology , Gene Expression Regulation , Hearing Loss, Sensorineural/pathology , High-Throughput Nucleotide Sequencing , Homer Scaffolding Proteins , Humans , Mice , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Stereocilia/genetics , Stereocilia/pathology , Zebrafish , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
4.
Int J Audiol ; 52(12): 832-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24160897

ABSTRACT

OBJECTIVE: The purpose of this study was to compare the genotype/phenotype relationship between siblings with identical USH2A pathologic mutations and the consequent audiologic phenotypes, in particular degree of hearing loss (HL). Decade audiograms were also compared among two groups of affected subjects with different mutations of USH2A. DESIGN: DNA samples from patients with Usher syndrome type II were analysed. The audiological features of patients and affected siblings with USH2A mutations were also examined to identify genotype-phenotype correlations. STUDY SAMPLE: Genetic and audiometric examinations were performed in 18 subjects from nine families with Usher syndrome type IIA. RESULTS: Three different USH2A mutations were identified in the affected subjects. Both similarities and differences of the auditory phenotype were seen in families with several affected siblings. A variable degree of hearing loss, ranging from mild to profound, was observed among affected subjects. No significant differences in hearing thresholds were found the group of affected subjects with different pathological mutations. CONCLUSIONS: Our results indicate that mutations in the USH2A gene and the resulting phenotype are probably modulated by other variables, such as modifying genes, epigenetics or environmental factors which may be of importance for better understanding the etiology of Usher syndrome.


Subject(s)
Hearing , Usher Syndromes/physiopathology , Acoustic Impedance Tests , Adolescent , Adult , Aged , Audiometry, Pure-Tone , Auditory Threshold , Child , DNA Mutational Analysis , Extracellular Matrix Proteins/genetics , Female , Genetic Association Studies , Genetic Predisposition to Disease , Hearing/genetics , Humans , Male , Middle Aged , Mutation , Nebraska , Pedigree , Phenotype , Risk Factors , Severity of Illness Index , Sweden , Usher Syndromes/diagnosis , Usher Syndromes/genetics
5.
J Med Genet ; 50(9): 627-34, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23804846

ABSTRACT

BACKGROUND: Non-syndromic hearing loss (NSHL) is the most common sensory impairment in humans. Until recently its extreme genetic heterogeneity precluded comprehensive genetic testing. Using a platform that couples targeted genomic enrichment (TGE) and massively parallel sequencing (MPS) to sequence all exons of all genes implicated in NSHL, we tested 100 persons with presumed genetic NSHL and in so doing established sequencing requirements for maximum sensitivity and defined MPS quality score metrics that obviate Sanger validation of variants. METHODS: We examined DNA from 100 sequentially collected probands with presumed genetic NSHL without exclusions due to inheritance, previous genetic testing, or type of hearing loss. We performed TGE using post-capture multiplexing in variable pool sizes followed by Illumina sequencing. We developed a local Galaxy installation on a high performance computing cluster for bioinformatics analysis. RESULTS: To obtain maximum variant sensitivity with this platform 3.2-6.3 million total mapped sequencing reads per sample were required. Quality score analysis showed that Sanger validation was not required for 95% of variants. Our overall diagnostic rate was 42%, but this varied by clinical features from 0% for persons with asymmetric hearing loss to 56% for persons with bilateral autosomal recessive NSHL. CONCLUSIONS: These findings will direct the use of TGE and MPS strategies for genetic diagnosis for NSHL. Our diagnostic rate highlights the need for further research on genetic deafness focused on novel gene identification and an improved understanding of the role of non-exonic mutations. The unsolved families we have identified provide a valuable resource to address these areas.


Subject(s)
Deafness/genetics , Genetic Testing/methods , Genomics/methods , Adolescent , Adult , Female , Humans , Male , Polymorphism, Single Nucleotide , Reproducibility of Results , Sequence Analysis, DNA
6.
Mol Vis ; 18: 1719-26, 2012.
Article in English | MEDLINE | ID: mdl-22815625

ABSTRACT

PURPOSE: PCDH15 codes for protocadherin-15, a cell-cell adhesion protein essential in the morphogenesis and cohesion of stereocilia bundles and in the function or preservation of photoreceptor cells. Mutations in the PCDH15 gene are responsible for Usher syndrome type I (USH1F) and non-syndromic hearing loss (DFNB23). The purpose of this work was to perform PCDH15 mutation screening to identify the genetic cause of the disease in a cohort of Spanish patients with Usher syndrome type I and establish phenotype-genotype correlation. METHODS: Mutation analysis of PCDH15 included additional exons recently identified and was performed by direct sequencing. The screening was performed in 19 probands with USH already screened for mutations in the most prevalent USH1 genes, myosin VIIA (MYO7A) and cadherin-23 (CDH23), and for copy number variants in PCDH15. RESULTS: Seven different point mutations, five novel, were detected. Including the large PCDH15 rearrangements previously reported in our cohort of patients, a total of seven of 19 patients (36.8%) were carriers of at least one pathogenic allele. Thirteen out of the 38 screened alleles carried pathogenic PCDH15 variants (34.2%). CONCLUSIONS: Five out of the seven point mutations reported in the present study are novel, supporting the idea that most PCDH15 mutations are private. Furthermore, no mutational hotspots have been identified. In most patients, detected mutations led to a truncated protein, reinforcing the hypothesis that severe mutations cause the Usher I phenotype and that missense variants are mainly responsible for non-syndromic hearing impairment.


Subject(s)
Cadherins/genetics , Mutation , Usher Syndromes/genetics , White People/genetics , Alleles , Cadherin Related Proteins , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Exons , Female , Gene Dosage , Gene Frequency , Genetic Association Studies , Genotype , Heterozygote , Humans , Male , Pedigree , Spain , Young Adult
7.
Mol Vis ; 18: 1379-83, 2012.
Article in English | MEDLINE | ID: mdl-22690115

ABSTRACT

PURPOSE: To identify the genetic defect in a Hutterite population from northern Alberta with Usher syndrome type I. METHODS: Complete ophthalmic examinations were conducted on two boys and two girls from two related Hutterite families diagnosed with Usher syndrome type I. DNA from patients and their parents was first evaluated for a mutation in exon 10 of the protocadherin-related 15 (PCDH15) gene (c.1471delG), previously reported in southern Alberta Hutterite patients with Usher syndrome (USH1F). Single nucleotide polymorphic linkage analysis was then used to confirm another locus, and DNA was analyzed with the Usher Chip v4.0 platform. RESULTS: Severe hearing impairment, unintelligible speech, and retinitis pigmentosa with varying degrees of visual acuity and visual field loss established a clinical diagnosis of Usher syndrome type I. The patients did not carry the exon 10 mutation in the PCDH15 gene; however, with microarray analysis, a previously reported mutation (c.52C>T; p.Q18X) in the myosin VIIA (MYO7A) gene was found in the homozygous state in the affected siblings. CONCLUSIONS: The finding of a MYO7A mutation in two related Hutterite families from northern Alberta provides evidence of genetic heterogeneity in Hutterites affected by Usher syndrome type I.


Subject(s)
Cadherins/genetics , Ethnicity/genetics , Genetic Heterogeneity , Myosins/genetics , Usher Syndromes/genetics , Adolescent , Alberta , Cadherin Related Proteins , Child , Exons , Female , Genetic Linkage , Genotype , Homozygote , Humans , Male , Myosin VIIa , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Siblings , Usher Syndromes/pathology
8.
Invest Ophthalmol Vis Sci ; 52(11): 7924-36, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21873662

ABSTRACT

PURPOSE. To determine the disease course in Usher syndrome type IB (USH1B) caused by myosin 7A (MYO7A) gene mutations. METHODS. USH1B patients (n = 33, ages 2-61) representing 25 different families were studied by ocular examination, kinetic and chromatic static perimetry, dark adaptometry, and optical coherence tomography (OCT). Consequences of the mutant alleles were predicted. RESULTS. All MYO7A patients had severely abnormal ERGs, but kinetic fields revealed regional patterns of visual loss that suggested a disease sequence. Rod-mediated vision could be lost to different degrees in the first decades of life. Cone vision followed a more predictable and slower decline. Central vision ranged from normal to reduced in the first four decades of life and thereafter was severely abnormal. Dark adaptation kinetics was normal. Photoreceptor layer thickness in a wide region of central retina could differ dramatically between patients of comparable ages; and there were examples of severe losses in childhood as well as relative preservation in patients in the third decade of life. Comparisons were made between the mutant alleles in mild versus more severe phenotypes. CONCLUSIONS. A disease sequence in USH1B leads from generally full but impaired visual fields to residual small central islands. At most disease stages, there was preserved temporal peripheral field, a potential target for early phase clinical trials of gene therapy. From data comparing patients' rod disease in this cohort, the authors speculate that null MYO7A alleles could be associated with milder dysfunction and fewer photoreceptor structural losses at ages when other genotypes show more severe phenotypes.


Subject(s)
Mutation , Myosins/genetics , Retinal Degeneration/physiopathology , Usher Syndromes/physiopathology , Vision Disorders/physiopathology , Visual Fields/physiology , Adolescent , Adult , Child , Child, Preschool , Dark Adaptation , Electroretinography , Female , Humans , Male , Middle Aged , Myosin VIIa , Phenotype , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Tomography, Optical Coherence , Usher Syndromes/genetics , Vision Disorders/genetics , Visual Field Tests , Young Adult
9.
Adv Otorhinolaryngol ; 70: 75-83, 2011.
Article in English | MEDLINE | ID: mdl-21358188

ABSTRACT

There are several syndromes in which both hearing and renal function are impaired. The two best known are branchio-oto-renal (BOR) syndrome and Alport syndrome. These are reviewed along with several other rarer syndromes. BOR is especially important since it is likely to be first recognized by the otolaryngologist because of the hearing and branchial anomalies. It is important for the practicing otolaryngologist to recognize these disorders and to ensure that renal problems are being treated. In addition, the syndromes discussed here are all hereditary and referral to a clinical geneticist may be helpful to the individual and family.


Subject(s)
Branchio-Oto-Renal Syndrome/genetics , Nephritis, Hereditary/genetics , Branchio-Oto-Renal Syndrome/diagnosis , Branchio-Oto-Renal Syndrome/therapy , Collagen Type IV/genetics , Diagnosis, Differential , Genetic Counseling , Heredity , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Nephritis, Hereditary/diagnosis , Nephritis, Hereditary/therapy , Nuclear Proteins/genetics , Protein Tyrosine Phosphatases/genetics , Referral and Consultation
10.
Am J Hum Genet ; 88(2): 127-37, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21255762

ABSTRACT

By using homozygosity mapping in a consanguineous Pakistani family, we detected linkage of nonsyndromic hearing loss to a 7.6 Mb region on chromosome 3q13.31-q21.1 within the previously reported DFNB42 locus. Subsequent candidate gene sequencing identified a homozygous nonsense mutation (c.1135G>T [p.Glu379X]) in ILDR1 as the cause of hearing impairment. By analyzing additional consanguineous families with homozygosity at this locus, we detected ILDR1 mutations in the affected individuals of 10 more families from Pakistan and Iran. The identified ILDR1 variants include missense, nonsense, frameshift, and splice-site mutations as well as a start codon mutation in the family that originally defined the DFNB42 locus. ILDR1 encodes the evolutionarily conserved immunoglobulin-like domain containing receptor 1, a putative transmembrane receptor of unknown function. In situ hybridization detected expression of Ildr1, the murine ortholog, early in development in the vestibule and in hair cells and supporting cells of the cochlea. Expression in hair cell- and supporting cell-containing neurosensory organs is conserved in the zebrafish, in which the ildr1 ortholog is prominently expressed in the developing ear and neuromasts of the lateral line. These data identify loss-of-function mutations of ILDR1, a gene with a conserved expression pattern pointing to a conserved function in hearing in vertebrates, as underlying nonsyndromic prelingual sensorineural hearing impairment.


Subject(s)
Codon, Nonsense/genetics , Genes, Recessive/genetics , Genetic Predisposition to Disease , Hearing Loss/genetics , Receptors, Cell Surface/genetics , Animals , Chromosome Mapping , Chromosomes, Human, Pair 3/genetics , Consanguinity , Ear, Inner , Female , Genetic Linkage , Genotype , Humans , In Situ Hybridization , Lod Score , Male , Mice , Pedigree , Zebrafish
11.
Eur J Ophthalmol ; 21(1): 30-8, 2011.
Article in English | MEDLINE | ID: mdl-20544672

ABSTRACT

PURPOSE: To evaluate the retinal function, with emphasis on phenotype and rate of progression, in infants and children with different genotypes of Usher syndrome. METHODS: Fourteen children (2-10 years of age) with retinitis pigmentosa and hearing impairment were examined with full-field electroretinography (ERG) during general anesthesia, ophthalmologic examination, and genetic analysis. Five children were repeatedly examined (follow-up 5-10 years) with full-field ERG under local anesthesia and in 2 children multifocal ERG and optical coherence tomography (OCT) were performed. These results were compared to full-field ERG data from 58 children without retinal eye disorder. RESULTS: Six children were genotyped as Usher 1B, 2A, and 3A. Full-field ERG demonstrated early alterations corresponding to a rod-cone dystrophy in all children. A remaining rod function could be verified in the majority of the children up to 4 years of age. After 4 years of age, there was a further deterioration of the rod function; the progress was severe in Usher types 1 and 2 and moderate in Usher type 3. In all children, the cone function was moderately reduced, in a few cases almost normal. The results from the 58 children without retinal disorder confirm that full-field ERG during general anesthesia is reliable. Multifocal ERG confirmed a preserved central cone function and in OCT there were discrete structural alterations. CONCLUSIONS: Full-field ERG during general anesthesia in children with Usher syndrome demonstrates variable phenotypes and different degrees in rate of progression during childhood.


Subject(s)
Photoreceptor Cells, Vertebrate/physiology , Usher Syndromes/physiopathology , Anesthesia, General , Child , Child, Preschool , Disease Progression , Electroretinography , Follow-Up Studies , Genotype , Humans , Phenotype , Registries , Retinitis Pigmentosa/physiopathology , Tomography, Optical Coherence , Usher Syndromes/genetics
12.
Ophthalmic Genet ; 32(2): 65-74, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21174530

ABSTRACT

OBJECTIVE: To characterize visual function in defined genotypes including siblings with Usher syndrome. METHODS: Thirteen patients with phenotypically different subtypes of Usher syndrome, including 3 families with affected siblings, were selected. Genetic analysis and ophthalmological examinations including visual fields, full-field electroretinography (ERG), multifocal electroretinography (mf ERG), and optical coherence tomography (OCT) were assessed. The patients' degree of visual handicap was evaluated by a questionnaire (ADL). RESULTS: Twelve of thirteen patients were genotyped as Usher 1B, 1D, 1F, 2A, 2C or 3A. In 12 of 13 patients examined with ERG the 30 Hz flickering light response revealed remaining cone function. In 3 of the patients with Usher type 1 mf ERG demonstrated a specific pattern, with a sharp distinction between the area with reduced function and the central area with remaining macular function and normal peak time. OCT demonstrated loss of foveal depression with distortion of the foveal architecture in the macula in all patients. The foveal thickness ranged from 159 to 384 µm and was not correlated to retinal function. Three siblings shared the same mutation for Usher 2C but in contrast to previous reports regarding this genotype, 1 of them diverged in phenotype with substantially normal visual fields, almost normal OCT and mf ERG findings, and only moderately reduced rod and cone function according to ERG. CONCLUSIONS: Evaluation of visual function comprising both the severity of the rod cone degeneration and the function in the macular region confirm phenotypical heterogeneity within siblings and between different genotypes of Usher syndrome.


Subject(s)
Genotype , Phenotype , Usher Syndromes/genetics , Adolescent , Adult , Aged , Cadherin Related Proteins , Cadherins/genetics , Child , Electroretinography , Extracellular Matrix Proteins/genetics , Female , Humans , Male , Membrane Proteins/genetics , Middle Aged , Myosins/genetics , Pedigree , Receptors, G-Protein-Coupled/genetics , Retina/physiopathology , Siblings , Surveys and Questionnaires , Tomography, Optical Coherence , Usher Syndromes/physiopathology , Visual Acuity/physiology , Visual Fields/physiology
13.
Laryngoscope ; 120(12): 2489-93, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21046548

ABSTRACT

OBJECTIVES/HYPOTHESIS: To determine the cause of autosomal dominant hearing loss segregating in an American family. STUDY DESIGN: Family study. METHODS: Otologic and audiometric examination was performed on affected family members. Genome wide parametric multipoint linkage mapping using a dominant model was performed with Affymetrix 50K GeneChip data. Direct sequencing was used to confirm the causative mutation. RESULTS: In American family 467, segregating autosomal dominant nonsyndromic hearing loss, a novel heterozygous missense mutation (c.362T>C; p.F121S) was identified in the COCH gene. This mutation was also associated with vestibular dysfunction typical of other DFNA9 families. However, affected family members also exhibited memory loss and night blindness. CONCLUSIONS: The novel COCH mutation affects the functionally important limulus factor C, Coch-5b2 and Lgl1 domain where most DFNA9 mutations have been localized. The onset of the hearing loss, in the 2nd or 3rd decade of life, is earlier than in most DFNA9 families. The progression of hearing loss and vestibular dysfunction in the American family is typical of other DFNA9 families with mutations in this domain. Memory loss and night blindness have not been previously reported in DFNA9 families.


Subject(s)
DNA/genetics , Genetic Association Studies/methods , Genetic Predisposition to Disease , Hearing Loss, Sensorineural/genetics , Mutation, Missense , Proteins/genetics , Audiometry , Disease Progression , Extracellular Matrix Proteins , Family , Female , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/physiopathology , Humans , Male , Pedigree , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , United States
14.
Genet Med ; 12(8): 512-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20613545

ABSTRACT

PURPOSE: Usher syndrome is a major cause of genetic deafness and blindness. The hearing loss is usually congenital and the retinitis pigmentosa is progressive and first noticed in early childhood to the middle teenage years. Its frequency may be underestimated. Newly developed molecular technologies can detect the underlying gene mutation of this disorder early in life providing estimation of its prevalence in at risk pediatric populations and laying a foundation for its incorporation as an adjunct to newborn hearing screening programs. METHODS: A total of 133 children from two deaf and hard of hearing pediatric populations were genotyped first for GJB2/6 and, if negative, then for Usher syndrome. Children were scored as positive if the test revealed > or =1 pathogenic mutations in any Usher gene. RESULTS: Fifteen children carried pathogenic mutations in one of the Usher genes; the number of deaf and hard of hearing children carrying Usher syndrome mutations was 15/133 (11.3%). The population prevalence was estimated to be 1/6000. CONCLUSION: Usher syndrome is more prevalent than has been reported before the genome project era. Early diagnosis of Usher syndrome has important positive implications for childhood safety, educational planning, genetic counseling, and treatment. The results demonstrate that DNA testing for Usher syndrome is feasible and may be a useful addition to newborn hearing screening programs.


Subject(s)
Connexins/genetics , Genetic Testing/methods , Usher Syndromes/epidemiology , Usher Syndromes/genetics , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Cadherin Related Proteins , Cadherins/genetics , Cell Cycle Proteins , Connexin 26 , Connexin 30 , Cytoskeletal Proteins , Extracellular Matrix Proteins/genetics , Female , Humans , Male , Microarray Analysis , Mutation/genetics , Myosin VIIa , Myosins/genetics , Oregon/epidemiology , Prevalence , Sequence Analysis, DNA , Surveys and Questionnaires , Young Adult
15.
Ann Otol Rhinol Laryngol ; 119(12): 830-5, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21250555

ABSTRACT

OBJECTIVES: We investigated the cause of autosomal recessive nonsyndromic hearing loss (ARNSHL) that segregated in 2 consanguineous Iranian families. METHODS: Otologic and audiometric examinations were performed on affected members of each family. Genome-wide parametric multipoint linkage mapping using a recessive model was performed with Affymetrix 50K GeneChips or short tandem repeat polymorphisms. Direct sequencing was used to confirm the causative mutation in each family. RESULTS: In 2 Iranian families, L-1651 and L-8600606, with ARNSHL that mapped to the DFNB7/11 locus, homozygosity for a reported splice site mutation (c.776+1G>A), and a novel deletion (c.1589_1590delCT; p.S530*) were identified in the TMC1 gene, respectively. CONCLUSIONS: Consistent with the previously reported phenotype in DFNB7/11 families, the 2 Iranian families had segregated congenital, profound hearing impairment. However, in family L-1651, one affected family member (IV:3) has milder hearing impairment than expected, suggesting a potential genetic modifier effect. These results indicate that DFNB7/11 is a common form of genetic hearing loss in Iran, because this population is the source of 6 of the 29 TMC1 mutations reported worldwide.


Subject(s)
Hearing Loss/genetics , Membrane Proteins/genetics , Mutation , Chromosome Mapping , Computers, Handheld , Consanguinity , Deafness/congenital , Deafness/genetics , Genome-Wide Association Study , Genotype , Hearing Loss/congenital , Humans , Iran , Microsatellite Repeats , Pedigree , Polymorphism, Single Nucleotide , RNA Splice Sites/genetics , Sequence Analysis, DNA , Sequence Deletion
16.
Am J Hum Genet ; 85(3): 328-37, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19732867

ABSTRACT

Hearing loss is the most common form of sensory impairment in humans and is frequently progressive in nature. Here we link a previously uncharacterized gene to hearing impairment in mice and humans. We show that hearing loss in the ethylnitrosourea (ENU)-induced samba mouse line is caused by a mutation in Loxhd1. LOXHD1 consists entirely of PLAT (polycystin/lipoxygenase/alpha-toxin) domains and is expressed along the membrane of mature hair cell stereocilia. Stereociliary development is unaffected in samba mice, but hair cell function is perturbed and hair cells eventually degenerate. Based on the studies in mice, we screened DNA from human families segregating deafness and identified a mutation in LOXHD1, which causes DFNB77, a progressive form of autosomal-recessive nonsyndromic hearing loss (ARNSHL). LOXHD1, MYO3a, and PJVK are the only human genes to date linked to progressive ARNSHL. These three genes are required for hair cell function, suggesting that age-dependent hair cell failure is a common mechanism for progressive ARNSHL.


Subject(s)
Carrier Proteins/genetics , Conserved Sequence , Evolution, Molecular , Hair Cells, Auditory, Outer/pathology , Hearing Loss/genetics , Mutation/genetics , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/chemistry , Cilia/pathology , Cilia/ultrastructure , Codon, Terminator/genetics , DNA Mutational Analysis , Genes, Recessive , Hair Cells, Auditory, Outer/ultrastructure , Hearing Loss/pathology , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , In Situ Hybridization , Mice , Molecular Sequence Data , Mutation, Missense/genetics , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Protein Structure, Secondary , Spiral Ganglion/pathology , Spiral Ganglion/ultrastructure
17.
Laryngoscope ; 119(4): 727-33, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19274735

ABSTRACT

OBJECTIVES: To use clinical and genetic analyses to determine the mutation causing autosomal recessive nonsyndromic hearing loss (ARNSHL) segregating in two consanguineous Iranian families. STUDY DESIGN: Family study. METHODS: Members of each family received otologic and audiometric examination for the type and extent of hearing loss. Linkage mapping using Affymetrix 50K GeneChips and short tandem repeat (STRP) analysis localized the hearing loss in both families to the DFNB3 locus. Direct sequencing of the MYO15A gene was completed on affected members of both families. RESULTS: Family L-3165 segregated a novel homozygous missense mutation (c.6371G>A) that results in a p.R2124Q amino acid substitution in the myosin XVa protein, while family L-896 segregated a novel homozygous missense (c.6555C>T) mutation resulting in a p.P2073S amino acid change. CONCLUSIONS: These are the first MYO15A mutations reported to cause DFNB3 sensorineural hearing loss in the Iranian population. Like other mutations located in the myosin tail homology 4 (MyTH4) domain, the p.R2124Q and p.P2073S mutations are predicted to disrupt the function of the myosin XVa protein, which is integral to the mechanosensory activity of hair cells in the inner ear.


Subject(s)
Chromosomes, Human, Pair 17/genetics , Hearing Loss, Sensorineural/genetics , Mutation, Missense , Myosins/genetics , Chromosome Mapping , Consanguinity , Female , Genotype , Humans , Iran , Lod Score , Male , Pedigree , Sequence Analysis, Protein
19.
Invest Ophthalmol Vis Sci ; 50(4): 1886-94, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19074810

ABSTRACT

PURPOSE: To study retinal microstructure in Usher Syndrome type 1B (USH1B) caused by MYO7A mutations as a prelude to treatment initiatives. METHODS: Patients with MYO7A-USH1B (n=17; ages 5-61) were studied with optical coherence tomography. Retinal laminae across horizontal and vertical meridians were measured. Colocalized visual sensitivity was measured with automated perimetry to enable comparisons of function and structure in the transition zones. RESULTS: Laminar architecture of the central retina in MYO7A-USH1B ranged from normal to severely abnormal. Within the transition zone between normal and abnormal retina, the first detectable abnormality was an increase in prominence of the OLM (outer limiting membrane). Declining ONL thickness was accompanied by increased thickness of the OPL and normal or increased INL. Undetectable ONL and OPL and hyperthick INL were features of severe laminopathy at further eccentricities into the transition zone. Visual sensitivity in the transition zone declined with the decrease in ONL thickness. CONCLUSIONS: Patients with MYO7A-USH1B can have regions of structurally and functionally normal retina with definable transitions to severe laminopathy and visual loss. The earliest detectable structural markers of disease may represent Müller glial cell response to photoreceptor stress and apoptosis. Visual losses were predictably related to a decline in ONL thickness. The prospect of focal treatment of MYO7A-USH1B, such as subretinal gene therapy, prompts the need to identify retinal locations that warrant consideration for treatment in early phase trials. The transition zones are candidate sites for treatment, and laminar architecture and visual sensitivity are possible outcomes to assess safety and efficacy.


Subject(s)
Mutation , Myosins/genetics , Retina/pathology , Retinal Diseases/genetics , Usher Syndromes/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Myosin VIIa , Retinal Diseases/pathology , Tomography, Optical Coherence , Usher Syndromes/pathology , Visual Acuity/physiology , Visual Field Tests , Visual Fields/physiology , Young Adult
20.
Eur J Hum Genet ; 17(4): 474-82, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18854872

ABSTRACT

Autosomal recessive retinitis pigmentosa (ARRP) is a genetically heterogeneous disorder. ARRP could be associated with extraocular manifestations that define specific syndromes such as Usher syndrome (USH) characterized by retinal degeneration and congenital hearing loss (HL). The USH type II (USH2) associates RP and mild-to-moderate HL with preserved vestibular function. At least three genes USH2A, the very large G-protein-coupled receptor, GPR98, and DFNB31 are responsible for USH2 syndrome. Here, we report on the segregation of non-syndromic ARRP and USH2 syndrome in a consanguineous Tunisian family, which was previously used to define USH2B locus. With regard to the co-occurrence of these two different pathologies, clinical and genetic reanalysis of the extended family showed (i) phenotypic heterogeneity within USH2 patients and (ii) excluded linkage to USH2B locus. Indeed, linkage analysis disclosed the cosegregation of the USH2 phenotype with the USH2C locus markers, D5S428 and D5S618, whereas the ARRP perfectly segregates with PDE6B flanking markers D4S3360 and D4S2930. Molecular analysis revealed two new missense mutations, p.Y6044C and p.W807R, occurring in GPR98 and PDE6B genes, respectively. In conclusion, our results show that the USH2B locus at chromosome 3p23-24.2 does not exist, and we therefore withdraw this locus designation. The combination of molecular findings for GPR98 and PDE6B genes enable us to explain the phenotypic heterogeneity and particularly the severe ocular affection first observed in one USH2 patient. This report presents an illustration of how consanguinity could increase familial clustering of multiple hereditary diseases within the same family.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Mutation , Receptors, G-Protein-Coupled/genetics , Amino Acid Sequence , Base Sequence , Consanguinity , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Family , Hearing Loss/congenital , Humans , Molecular Sequence Data , Pedigree , Phenotype , Receptors, G-Protein-Coupled/metabolism , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Tunisia , Usher Syndromes/genetics , Usher Syndromes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL