Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
BMC Res Notes ; 17(1): 131, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730444

ABSTRACT

OBJECTIVES: The crystal structure of the six protomers of gap junction protein beta 2 (GJB2) enables prediction of the effect(s) of an amino acid substitution, thereby facilitating investigation of molecular pathogenesis of missense variants of GJB2. This study mainly focused on R143W variant that causes hearing loss, and investigated the relationship between amino acid substitution and 3-D structural changes in GJB2. METHODS: Patients with nonsyndromic hearing loss who appeared to have two GJB2 pathogenic variants, including the R143W variant, were investigated. Because the X-ray crystal structure of the six protomers of the GJB2 protein is known, R143W and structurally related variants of GJB2 were modeled using this crystal structure as a template. The wild-type crystal structure and the variant computer-aided model were observed and the differences in molecular interactions within the two were analyzed. RESULTS: The predicted structure demonstrated that the hydrogen bond between R143 and N206 was important for the stability of the protomer structure. From this prediction, R143W related N206S and N206T variants showed loss of the hydrogen bond. CONCLUSION: Investigation of the genotypes and clinical data in patients carrying the R143W variant on an allele indicated that severity of hearing loss depends largely on the levels of dysfunction of the pathogenic variant on the allele, whereas a patient with the homozygous R143W variant demonstrated profound hearing loss. We concluded that these hearing impairments may be due to destabilization of the protomer structure of GJB2 caused by the R143W variant.


Subject(s)
Connexin 26 , Connexins , Hearing Loss , Humans , Connexin 26/genetics , Connexins/genetics , Connexins/chemistry , Hearing Loss/genetics , Female , Male , Child , Models, Molecular , Child, Preschool , Mutation, Missense , Amino Acid Substitution , Hydrogen Bonding , Crystallography, X-Ray , Adolescent , Adult
2.
Neurosci Lett ; 803: 137178, 2023 04 23.
Article in English | MEDLINE | ID: mdl-36914046

ABSTRACT

Otof, which encodes otoferlin, knockout mice are considered model mice for auditory neuropathy spectrum disorder, which is characterized by an absent auditory brainstem response (ABR) despite preserved distortion product otoacoustic emission (DPOAE). Although otoferlin-deficient mice lack neurotransmitter release at the inner hair cell (IHC) synapse, it remains unclear how the Otof mutation affects spiral ganglions. Thus, we used Otof-mutant mice carrying the Otoftm1a(KOMP)Wtsi allele (Otoftm1a) and analyzed spiral ganglion neurons (SGNs) in Otoftm1a/tm1a mice by immunolabeling type Ⅰ SGNs (SGN-Ⅰ) and type II SGNs (SGN-II). We also examined apoptotic cells in SGNs. Four-week-old Otoftm1a/tm1a mice had an absent ABR but normal DPOAEs. The number of SGNs was significantly lower in Otoftm1a/tm1a mice on postnatal day 7 (P7), P14, and P28 compared with that of wild-type mice. Moreover, significantly more apoptotic SGNs were observed in Otoftm1a/tm1a mice than in wild-type mice on P7, P14, and P28. SGN-IIs were not significantly reduced in Otoftm1a/tm1a mice on P7, P14, and P28. No apoptotic SGN-IIs were observed under our experimental conditions. In summary, Otoftm1a/tm1a mice showed a reduction in SGNs accompanied by apoptosis of SGN-Ⅰs even before the onset of hearing. We speculate that the reduction in SGNs with apoptosis is a secondary defect caused by a lack of otoferlin in IHCs. Appropriate glutamatergic synaptic inputs may be important for the survival of SGNs.


Subject(s)
Neurons , Spiral Ganglion , Animals , Mice , Spiral Ganglion/metabolism , Neurons/metabolism , Apoptosis/physiology , Synaptic Transmission/physiology , Mice, Knockout , Membrane Proteins/genetics , Membrane Proteins/metabolism
3.
Otol Neurotol ; 41(6): e663-e673, 2020 07.
Article in English | MEDLINE | ID: mdl-32097363

ABSTRACT

HYPOTHESIS: The phenotype of DFNA11 consists of specific features at diverse developmental and age stages. BACKGROUND: Only eight mutations have been identified for autosomal dominant non-syndromic hearing loss related to MYO7A (DFNA11), and the onset and progression of DFNA11 are poorly understood. METHODS: After linkage analysis and following Sanger sequencing in a family suspected to have autosomal dominant hereditary hearing loss, we analyzed the audiometric and vestibular functions and their long-term changes in the subjects carrying the variant. RESULTS: A reported variant of uncertain significance, NP_000251.3:p.Arg853His, in MYO7A was detected and cosegregation data of this large family provided evidence that the variant was likely pathogenic for DFNA11. Family members with the variant had no other symptoms associated with hearing loss and were confirmed to have autosomal dominant non-syndromic sensorineural hearing loss. Audiograms tended to show gently sloping configuration in childhood and flat configuration after the age of 30 years. Hearing loss at high frequencies progressed slowly, while hearing at low frequencies started to deteriorate later but progressed more rapidly. Some subjects showed partly abnormal results in the distortion products of otoacoustic emissions before the elevation of hearing thresholds. Vestibular function was within the normal range in all the subjects tested. CONCLUSION: We revealed that hearing loss at high frequencies was mainly noted in early developmental stages and that thresholds increased more rapidly in the low frequency range, resulting in changes in audiometric configuration. Deterioration of distortion product otoacoustic emissions (DPOAE) before the elevation of hearing thresholds was considered as a clinical feature of DFNA11.


Subject(s)
Hearing Loss, Sensorineural , Myosin VIIa/genetics , Adult , Aging , Child , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/genetics , Humans , Myosins/genetics , Otoacoustic Emissions, Spontaneous , Pedigree
4.
Int J Pediatr Otorhinolaryngol ; 130: 109831, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31875531

ABSTRACT

BACKGROUND: Hereditary hearing loss is characterized by a very high genetic heterogeneity. The OTOF (Locus: DFNB9), encoding otoferlin, is reported to be one of the major causes of non-syndromic hearing loss, and is also reported to be the most common cause of non-syndromic recessive auditory neuropathy spectrum disorder. METHODS: In this study, whole exome sequencing was employed for detection of novel pathogenic variant that segregates with autosomal recessive nonsyndromic hearing loss in a tribal family from Rajouri, Jammu and Kashmir. Proband was a 9-year-old male born to first-cousin parents and presented with sensorineural hearing loss since birth. Family resides in an area with high consanguinity and lack of basic health care facilities including genetic counselling services. RESULTS: We report a novel OTOF pathogenic variant NM_194248.2:c.4249_4250insG (p.Ser1417CysfsTer4) co-segregating with hearing loss in this family and not present in any public databases. CONCLUSIONS: Our findings not only extend the geographical and mutational spectrum of autosomal recessive nonsyndromic hearing loss but also support the need for introducing genetic counselling services to rural and tribal areas in India with high consanguinity.


Subject(s)
Deafness/genetics , Membrane Proteins/genetics , Mutation/genetics , Child , Consanguinity , Female , Humans , India , Male , Pedigree , Exome Sequencing
5.
Ear Hear ; 40(1): 184-191, 2019.
Article in English | MEDLINE | ID: mdl-29688962

ABSTRACT

OBJECTIVES: Auditory neuropathy (AN) is a clinical disorder characterized by the absence of auditory brainstem response and presence of otoacoustic emissions. A gradual loss of otoacoustic emissions has been reported for some cases of AN. Such cases could be diagnosed as cochlear hearing loss and lead to misunderstanding of the pathology when patients first visit clinics after the loss of otoacoustic emissions. The purpose of this study was to investigate the time course of changes in distortion product otoacoustic emissions (DPOAEs) in association with patients' genetic and clinical backgrounds, including the use of hearing aids. DESIGN: DPOAE measurements from 31 patients with AN were assessed. Genetic analyses for GJB2, OTOF, and mitochondrial m.1555A> G and m.3243A> G mutations were conducted for all cases, and the analyses for CDH23 and OPA1 were conducted for the selected cases. Patients who were younger than 10 years of age at the time of AN diagnosis were designated as the pediatric AN group (22 cases), and those who were 18 years of age or older were designated as the adult AN group (9 cases). DPOAE was measured at least twice in all patients. The response rate for DPOAEs was defined and analyzed. RESULTS: The pediatric AN group comprised 10 patients with OTOF mutations, 1 with GJB2 mutations, 1 with OPA1 mutation, and 10 with indefinite causes. Twelve ears (27%) showed no change in DPOAE, 20 ears (46%) showed a decrease in DPOAE, and 12 ears (27%) lost DPOAE. Loss of DPOAE occurred in one ear (2%) at 0 years of age and four ears (9%) at 1 year of age. The time courses of DPOAEs in patients with OTOF mutations were divided into those with early loss and those with no change, indicating that the mechanism for deterioration of DPOAEs includes not only the OTOF mutations but also other common modifier factors. Most, but not all, AN patients who used hearing aids showed deterioration of DPOAEs after the start of using hearing aids. A few AN patients also showed deterioration of DPOAEs before using hearing aids. The adult AN group comprised 2 patients with OPA1 mutations, 2 with OTOF mutations, and 5 with indefinite causes. Four ears (22%) showed no change in DPOAE, 13 ears (72%) showed a decrease, and one ear (6%) showed a loss of DPOAE. Although the ratio of DPOAE decrease was higher in the adult AN group than in the pediatric AN group, the ratio of DPOAE loss was lower in the adult AN group. DPOAE was not lost in all four ears with OPA1 mutations and in all four ears with OTOF mutations in the adult group. CONCLUSIONS: DPOAE was decreased or lost in approximately 70% of pediatric and about 80% of adult AN patients. Eleven percent of pediatric AN patients lost DPOAEs by 1 year of age. Genetic factors were thought to have influenced the time course of DPOAEs in the pediatric AN group. In most adult AN patients, DPOAE was rarely lost regardless of the genetic cause.


Subject(s)
Hearing Loss, Central/physiopathology , Otoacoustic Emissions, Spontaneous/physiology , Adolescent , Adult , Aged , Cadherin Related Proteins , Cadherins/genetics , Child , Child, Preschool , Connexin 26 , Connexins/genetics , Diagnostic Errors , Disease Progression , Female , GTP Phosphohydrolases/genetics , Genes, Mitochondrial/genetics , Hearing Loss, Central/diagnosis , Hearing Loss, Central/genetics , Hearing Loss, Sensorineural/diagnosis , Humans , Infant , Male , Membrane Proteins/genetics , Middle Aged , Retrospective Studies , Young Adult
6.
Auris Nasus Larynx ; 45(2): 222-226, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28502583

ABSTRACT

OBJECTIVE: To examine and expand the genetic spectrum of Waardenburg syndrome type 1 (WS1). METHODS: Clinical features related to Waardenburg syndrome (WS) were examined in a five-year old patient. Mutation analysis of genes related to WS was performed in the proband and her parents. Molecular modeling of EDNRB and the p.R319W mutant was conducted to predict the pathogenicity of the mutation. RESULTS: The proband showed sensorineural hearing loss, heterochromia iridis, and dystopia canthorum, fulfilling the clinical criteria of WS1. Genetic analyses revealed that the proband had no mutation in PAX3 which has been known as the cause of WS1, but had a homozygous missense mutation (p.R319W) in endothelin receptor type B (EDNRB) gene. The asymptomatic parents had the mutation in a heterozygote state. This mutation has been previously reported in a heterozygous state in a patient with Hirschsprung's disease unaccompanied by WS, but the patient and her parents did not show any symptoms in gastrointestinal tract. Molecular modeling of EDNRB with the p.R319W mutation demonstrated reduction of the positively charged surface area in this region, which might reduce binding ability of EDNRB to G protein and lead to abnormal signal transduction underlying the WS phenotype. CONCLUSIONS: Our findings suggested that autosomal recessive mutation in EDNRB may underlie a part of WS1 with the current diagnostic criteria, and supported that Hirschsprung's disease is a multifactorial genetic disease which requires additional factors. Further molecular analysis is necessary to elucidate the gene interaction and to reappraise the current WS classification.


Subject(s)
Receptor, Endothelin B/genetics , Waardenburg Syndrome/genetics , Child, Preschool , Female , Homozygote , Humans , Models, Molecular , Mutation, Missense , Waardenburg Syndrome/physiopathology
7.
Orphanet J Rare Dis ; 12(1): 157, 2017 09 25.
Article in English | MEDLINE | ID: mdl-28946916

ABSTRACT

BACKGROUND: To date, 102 genes have been reported as responsible for non-syndromic hearing loss, some of which are associated with specific audiogram features. Four genes have been reported as causative for mid-frequency sensorineural hearing loss (MFSNHL), among which TECTA is the most frequently reported; however, the prevalence of TECTA mutations is unknown. To elucidate the prevalence of TECTA mutation in MFSNHL and clarify genotype-phenotype correlations, we analyzed the genetic and clinical features of patients with MFSNHL. METHODS: Subjects with bilateral non-syndromic hearing loss were prescreened for GJB2 and m.1555A > G and m.3243A > G mitochondrial DNA mutations, and patients with inner ear malformations were excluded. We selected MFSNHL patients whose audiograms met the U-shaped criterion proposed by the GENDEAF study group, along with those with shallow U-shaped audiograms, for TECTA analysis. All TECTA exons were analyzed by Sanger sequencing. Novel missense variants were classified as possibly pathogenic, non-pathogenic, and variants of uncertain significance, based on genetic data. To evaluate novel possibly pathogenic variants, we predicted changes in protein structure by molecular modeling. RESULTS: Pathogenic and possibly pathogenic variants of TECTA were found in 4 (6.0%) of 67 patients with MFSNHL. In patients with U-shaped audiograms, none (0%) of 21 had pathogenic or possibly pathogenic variants. In patients with shallow U-shaped audiograms, four (8.7%) of 46 had pathogenic or possibly pathogenic variants. Two novel possibly pathogenic variants were identified and two previously reported mutations were considered as variant of unknown significance. The clinical features of patients with pathogenic and possibly pathogenic variants were consistent with those in previous studies. Pathogenic or possibly pathogenic variants were identified in 3 of 23 families (13.0%) which have the family histories compatible with autosomal dominant and 1 of 44 families (2.3%) which have the family histories compatible with sporadic or autosomal recessive. CONCLUSIONS: TECTA mutations were identified in 6.0% of MFSNHL. These mutations were more frequent in patients with shallow U-shaped audiograms than those with U-shaped audiograms, and in families which have the family histories compatible with autosomal dominant than those with the family histories compatible with sporadic or autosomal recessive.


Subject(s)
Extracellular Matrix Proteins/genetics , Hearing Loss, Sensorineural/genetics , DNA, Mitochondrial/genetics , Female , GPI-Linked Proteins/genetics , Humans , Male , Mutation , Pedigree
8.
Laryngoscope ; 127(7): 1663-1669, 2017 07.
Article in English | MEDLINE | ID: mdl-27577114

ABSTRACT

OBJECTIVES/HYPOTHESIS: To determine the frequency of the incomplete partition type III anomaly and the genetic and clinical features associated with POU3F4 mutations in children with hearing loss. STUDY DESIGN: Retrospective case series from 2000 to 2014 at the National Hospital Organization Tokyo Medical Center and collaborating hospitals. METHODS: A total of 1,004 patients (from 938 families) who had hearing loss by 10 years of age and had undergone computed tomography scanning of their temporal bones were enrolled in this genetic, clinical, and radiological study. RESULTS: The incomplete partition type III anomaly was identified in six patients (0.6%), each of whom had an enlargement of the vestibular aqueduct at the end close to the vestibule. The six patients also had POU3F4 variants, and a genetic analysis revealed frameshift deletions in three patients, a missense variant in two patients of the same family, and a large deletion in one patient. Three of the six patients with POU3F4 variants were sporadic cases, and in one patient the genetic mutation occurred de novo. CONCLUSIONS: It was indicated that POU3F4 mutations can be predicted by incomplete partition type III anomaly by radiological examination of the inner ear. All six of the patients showed mixed hearing loss, but none showed fluctuations in hearing, which may be related to the lack of vestibular aqueduct enlargement at the operculum. LEVEL OF EVIDENCE: 4 Laryngoscope, 127:1663-1669, 2017.


Subject(s)
Cochlea/abnormalities , DNA Mutational Analysis , Gene Frequency , Hearing Loss, Mixed Conductive-Sensorineural/diagnostic imaging , Hearing Loss, Mixed Conductive-Sensorineural/genetics , POU Domain Factors/genetics , Child , Child, Preschool , Cochlea/diagnostic imaging , Female , Humans , Male , Pedigree , Retrospective Studies , Tokyo , Tomography, X-Ray Computed , Vestibular Aqueduct/abnormalities , Vestibular Aqueduct/diagnostic imaging
9.
Auris Nasus Larynx ; 43(6): 609-13, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26973026

ABSTRACT

OBJECTIVE: To report two novel LOXHD1 mutations, including missense mutations and the clinical features of the patients. METHODS: We studied a three-generation Japanese family with hearing loss. Targeted next-generation sequencing was used for genetic analysis. Conditional orientation response audiometry and pure tone audiometry were used to assess hearing. SWISS-MODEL was used for molecular modeling of the PLAT domain in LOXHD1 protein. RESULTS: The two sisters, who had either mild or severe high-frequency hearing loss, were compound heterozygous for two novel mutations (c.5674G>T [p.V1892F] and c.4212+1G>A) in LOXHD1, which is responsible for autosomal-recessive nonsyndromic hearing loss DFNB77. These cases showed less severe hearing impairment than the previously reported cases carrying LOXHD1 mutations, but their hearing loss appeared to be progressive. Molecular modeling predicted that distorted structure of the PLAT domain in the p.V1892F mutant could lead to decreased affinity of the protein to lipid membrane resulting in hair cell dysfunction. CONCLUSION: We report a Japanese family carrying compound heterozygotes of truncating and nontruncating mutations in LOXHD1 identified by targeted NGS analysis. The fact of lower degree of hearing impairment in our cases than previously reported and the molecular modeling of the missense mutant provide insight to the genotype-phenotype correlation of DFNB77.


Subject(s)
Carrier Proteins/genetics , Hearing Loss, High-Frequency/genetics , Hearing Loss, Sensorineural/genetics , Mutation, Missense , Siblings , Asian People/genetics , Audiometry , Audiometry, Pure-Tone , Child , DNA Mutational Analysis , Female , Genotype , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Infant , Japan , Pedigree , Phenotype
10.
Otol Neurotol ; 37(4): 394-402, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26905822

ABSTRACT

HYPOTHESIS: Different missense mutations of the optic atrophy 1 gene (OPA1) identified in optic atrophy patients with auditory neuropathy spectrum disorder (ANSD) induce functional impairment through different molecular mechanisms. BACKGROUND: OPA1 is the gene responsible for autosomal dominant optic atrophy (ADOA), but some of its mutations are also associated with ANSD. OPA1 is a member of the GTPase family of proteins and plays a key role in the maintenance of mitochondrial activities that are dependent on dimer formation of the protein. There are many reports of OPA1 mutations, but the molecular mechanisms of their functional impairments are unclear. METHODS: The sequences of coding regions in OPA1 were analyzed from blood samples of ADOA patients with ANSD. Molecular modeling of the protein's ability to form dimers and its GTP-binding ability were conducted to study the effects of structural changes in OPA1 caused by two identified mutations and their resultant effects on protein function. RESULTS: Two heterozygous mutations, p.T414P (c.1240A>C) and p.T540P (c.1618A>C), located in the GTPase and middle domains of OPA1, respectively, were identified in two patients. Molecular modeling indicated decreased dimer formation caused by destabilization of the association structure of the p.T414P mutant, and decreased GTP-binding caused by destabilization of the binding site structure in the p.T540P mutant. CONCLUSION: These two different conformational changes might result in decreased GTPase activities that trigger ADOA associated with ANSD, and are likely to be associated with mild clinical features. Molecular modeling would provide useful information in clinical practice.


Subject(s)
GTP Phosphohydrolases/genetics , Hearing Loss, Central/genetics , Optic Atrophy, Autosomal Dominant/genetics , Adult , Female , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , Humans , Male , Models, Molecular , Mutation, Missense
11.
Orphanet J Rare Dis ; 10: 60, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25963016

ABSTRACT

BACKGROUND: Mutations in CDH23 are responsible for Usher syndrome 1D and recessive non-syndromic hearing loss. In this study, we revealed the prevalence of CDH23 mutations among patients with specific clinical characteristics. METHODS: After excluding patients with GJB2 mutations and mitochondrial m.1555A > G and m.3243A > G mutations, subjects for CDH23 mutation analysis were selected according to the following criteria: 1) Sporadic or recessively inherited hearing loss 2) bilateral non-syndromic congenital hearing loss, 3) no cochlear malformation, 4) a poorer hearing level at high frequencies than at low frequencies, and 5) severe or profound hearing loss at higher frequencies. RESULTS: Seventy-two subjects were selected from 621 consecutive probands who did not have environmental causes for their hearing loss. After direct sequencing, 13 of the 72 probands (18.1%) had homozygous or compound heterozygous CDH23 mutations. In total, we identified 16 CDH23 mutations, including five novel mutations. The 16 mutations included 12 missense, two frameshift, and two splice-site mutations. CONCLUSIONS: These results revealed that CDH23 mutations are highly prevalent in patients with congenital high-frequency sporadic or recessively inherited hearing loss and that the mutation spectrum was diverse, indicating that patients with these clinical features merit genetic analysis.


Subject(s)
Cadherins/genetics , Hearing Loss/genetics , Adolescent , Adult , Cadherin Related Proteins , Child , Child, Preschool , Connexin 26 , Connexins , DNA Mutational Analysis , Female , Hearing Loss, Sensorineural/genetics , Humans , Infant , Infant, Newborn , Male , Mutation/genetics , Phenotype , Prevalence , Young Adult
12.
Biochem Biophys Res Commun ; 447(3): 496-502, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24735539

ABSTRACT

The access of bone morphogenetic protein (BMP) to the BMP receptors on the cell surface is regulated by its antagonist noggin, which binds to heparan-sulfate proteoglycans on the cell surface. Noggin is encoded by NOG and mutations in the gene are associated with aberrant skeletal formation, such as in the autosomal dominant disorders proximal symphalangism (SYM1), multiple synostoses syndrome, Teunissen-Cremers syndrome, and tarsal-carpal coalition syndrome. NOG mutations affecting a specific function may produce a distinct phenotype. In this study, we investigated a Japanese pedigree with SYM1 and conductive hearing loss and found that it carried a novel heterozygous missense mutation of NOG (c.406C>T; p.R136C) affecting the heparin-binding site of noggin. As no mutations of the heparin-binding site of noggin have previously been reported, we investigated the crystal structure of wild-type noggin to investigate molecular mechanism of the p.R136C mutation. We found that the positively charged arginine at position 136 was predicted to be important for binding to the negatively charged heparan-sulfate proteoglycan (HSPG). An in silico docking analysis showed that one of the salt bridges between noggin and heparin disappeared following the replacement of the arginine with a non-charged cysteine. We propose that the decreased binding affinity of NOG with the p.R136C mutation to HSPG leads to an excess of BMP signaling and underlies the SYM1 and conductive hearing loss phenotype of carriers.


Subject(s)
Carrier Proteins/metabolism , Finger Joint/abnormalities , Hearing Loss, Conductive/genetics , Heparin/metabolism , Joint Diseases/congenital , Amino Acid Sequence , Amino Acid Substitution/genetics , Arginine/chemistry , Arginine/genetics , Asian People/genetics , Binding Sites/genetics , Bone Morphogenetic Protein 7/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Child , Crystallography, X-Ray , Cysteine/chemistry , Cysteine/genetics , Female , Heterozygote , Humans , Japan , Joint Diseases/genetics , Molecular Sequence Data , Mutation, Missense , Pedigree , Signal Transduction
13.
Gene ; 540(2): 258-62, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24582978

ABSTRACT

Waardenburg syndrome is characterized by hearing loss, pigmentation abnormalities, dysmorphologic features, and neurological phenotypes. Waardenburg syndrome consists of four distinct subtypes, and SOX10 mutations have been identified in type II and type IV. Type IV differs from type II owing to the presence of Hirschsprung disease. We identified a de novo nonsense mutation in SOX10 (p.G39X) in a female pediatric patient with Waardenburg syndrome with heterochromia iridis, profound bilateral sensorineural hearing loss, inner ear malformations, and overall hypopigmentation of the hair without dystopia canthorum. This patient has experienced chronic constipation since she was a neonate, but anorectal manometry showed a normal anorectal reflex. Chronic constipation in this patient was likely to be a consequence of a mild intestinal disorder owing to the SOX10 mutation, and this patient was considered to have a clinical phenotype intermediate between type II and type IV of the syndrome. Chronic constipation may be recognized as indicative of a SOX10 mutation in patients with Waardenburg syndrome.


Subject(s)
Codon, Nonsense , Constipation/diagnosis , SOXE Transcription Factors/genetics , Waardenburg Syndrome/diagnostic imaging , Base Sequence , Child, Preschool , Chronic Disease , Constipation/genetics , DNA Mutational Analysis , Female , Genetic Association Studies , Humans , Pedigree , Radiography , Temporal Bone/diagnostic imaging , Waardenburg Syndrome/genetics
14.
Orphanet J Rare Dis ; 8: 172, 2013 Oct 28.
Article in English | MEDLINE | ID: mdl-24164807

ABSTRACT

BACKGROUND: Genetic tests for hereditary hearing loss inform clinical management of patients and can provide the first step in the development of therapeutics. However, comprehensive genetic tests for deafness genes by Sanger sequencing is extremely expensive and time-consuming. Next-generation sequencing (NGS) technology is advantageous for genetic diagnosis of heterogeneous diseases that involve numerous causative genes. METHODS: Genomic DNA samples from 58 subjects with hearing loss from 15 unrelated Japanese families were subjected to NGS to identify the genetic causes of hearing loss. Subjects did not have pathogenic GJB2 mutations (the gene most often associated with inherited hearing loss), mitochondrial m.1555A>G or 3243A>G mutations, enlarged vestibular aqueduct, or auditory neuropathy. Clinical features of subjects were obtained from medical records. Genomic DNA was subjected to a custom-designed SureSelect Target Enrichment System to capture coding exons and proximal flanking intronic sequences of 84 genes responsible for nonsyndromic or syndromic hearing loss, and DNA was sequenced by Illumina GAIIx (paired-end read). The sequences were mapped and quality-checked using the programs BWA, Novoalign, Picard, and GATK, and analyzed by Avadis NGS. RESULTS: Candidate genes were identified in 7 of the 15 families. These genes were ACTG1, DFNA5, POU4F3, SLC26A5, SIX1, MYO7A, CDH23, PCDH15, and USH2A, suggesting that a variety of genes underlie early-childhood hearing loss in Japanese patients. Mutations in Usher syndrome-related genes were detected in three families, including one double heterozygous mutation of CDH23 and PCDH15. CONCLUSION: Targeted NGS analysis revealed a diverse spectrum of rare deafness genes in Japanese subjects and underscores implications for efficient genetic testing.


Subject(s)
Deafness/genetics , Hearing Loss/genetics , High-Throughput Nucleotide Sequencing/methods , Anion Transport Proteins/genetics , Asian People/genetics , Cadherin Related Proteins , Cadherins/genetics , Connexin 26 , Connexins , Cross-Sectional Studies , Genetic Testing , Hearing Loss, Sensorineural/genetics , Homeodomain Proteins/genetics , Humans , Myosin VIIa , Myosins/genetics , Sulfate Transporters , Transcription Factor Brn-3C/genetics , Usher Syndromes/genetics
15.
Biochem Biophys Res Commun ; 432(3): 475-9, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23399560

ABSTRACT

Genetic mutation is one of the causative factors for idiopathic progressive hearing loss. A patient with late-onset, moderate, and high-frequency hearing loss was found to have a novel, heterozygous KCNQ4 mutation, c.806_808delCCT, which led to a p.Ser260del located between S5 and the pore helix (PH). Molecular modeling analysis suggested that the p.Ser269del mutation could cause structural distortion and change in the electrostatic surface potential of the KCNQ4 channel protein, which may impede K+ transport. The present study supports the idea that a non-truncating mutation around the N-terminus of PH may be related to moderate hearing loss.


Subject(s)
Hearing Loss, High-Frequency/genetics , KCNQ Potassium Channels/chemistry , KCNQ Potassium Channels/genetics , Adult , Amino Acid Sequence , Female , Heterozygote , Humans , Models, Molecular , Molecular Sequence Data , Pedigree , Protein Structure, Secondary/genetics , Sequence Deletion , Serine/chemistry , Serine/genetics
16.
Methods Enzymol ; 521: 239-57, 2013.
Article in English | MEDLINE | ID: mdl-23351743

ABSTRACT

The formation of homo- and hetero-oligomers between various G protein-coupled receptors (GPCRs) has been demonstrated over the past decade. In most cases, GPCR heterodimerization increases the diversity of intracellular signaling. GPCR-type purinergic receptors (adenosine and P2Y receptors) are actively reported to form hetero-oligomers with each other, with GPCRs belonging to the same group (type 1, rhodopsin-like), and even with GPCRs from another group. This chapter describes common strategies to identify dimerization of purinergic receptors (coimmunoprecipitation, bioluminescence resonance energy transfer (BRET), and immunoelectron microscopy) and to assess the alteration of their pharmacology (ligand binding, intracellular cAMP, and intracellular Ca(2+) assays). We have reported dimerization of purinergic receptors using these strategies in transfected human embryonic kidney 293T cells and native brain tissue. Our data suggest that homo- and hetero-oligomerization between purinergic receptors exert unique pharmacology in this receptor group. According to these discoveries, heterodimerization is likely to be employed for the "fine-tuning" of purinergic receptor signaling.


Subject(s)
Receptors, Purinergic/analysis , Receptors, Purinergic/metabolism , HEK293 Cells , Humans , Immunoprecipitation/methods , Luminescent Measurements/methods , Microscopy, Electron/methods , Protein Multimerization , Signal Transduction
17.
Acta Otolaryngol ; 133(4): 345-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23163891

ABSTRACT

CONCLUSION: PAX3 genetic analysis increased the diagnostic accuracy for Waardenburg syndrome type I (WS1). Analysis of the three-dimensional (3D) structure of PAX3 helped verify the pathogenicity of a missense mutation, and multiple ligation-dependent probe amplification (MLPA) analysis of PAX3 increased the sensitivity of genetic diagnosis in patients with WS1. OBJECTIVES: Clinical diagnosis of WS1 is often difficult in individual patients with isolated, mild, or non-specific symptoms. The objective of the present study was to facilitate the accurate diagnosis of WS1 through genetic analysis of PAX3 and to expand the spectrum of known PAX3 mutations. METHODS: In two Japanese families with WS1, we conducted a clinical evaluation of symptoms and genetic analysis, which involved direct sequencing, MLPA analysis, quantitative PCR of PAX3, and analysis of the predicted 3D structure of PAX3. The normal-hearing control group comprised 92 subjects who had normal hearing according to pure tone audiometry. RESULTS: In one family, direct sequencing of PAX3 identified a heterozygous mutation, p.I59F. Analysis of PAX3 3D structures indicated that this mutation distorted the DNA-binding site of PAX3. In the other family, MLPA analysis and subsequent quantitative PCR detected a large, heterozygous deletion spanning 1759-2554 kb that eliminated 12-18 genes including a whole PAX3 gene.


Subject(s)
Genetic Predisposition to Disease , Paired Box Transcription Factors/genetics , Waardenburg Syndrome/diagnosis , Waardenburg Syndrome/genetics , Case-Control Studies , Child, Preschool , Female , Genetic Testing , Heterozygote , Humans , Infant , Japan , Male , Mutation, Missense , PAX3 Transcription Factor , Pedigree , Real-Time Polymerase Chain Reaction/methods
18.
BMC Res Notes ; 5: 145, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22420747

ABSTRACT

BACKGROUND: Mutation of the voltage-gated potassium channel KCNQ4 causes DFNA2-type nonsyndromic autosomal dominant sensorineural hearing loss. KCNQ4 is expressed predominantly in the auditory sensory outer hair cells, which are critical for sound amplification. RESULTS: We sequenced KCNQ4 from Japanese patients with sensorineural hearing loss, and identified a novel missense mutation encoding a Tyr270His located at the N-terminus of the highly conserved pore helix sequence. As this patient was not accessible to us and information about them was limited, we used molecular modeling to investigate whether this novel mutation is hypothetically pathogenic. A careful examination of an in silico structural model of the KCNQ4 pore region revealed that the Tyr270His mutation caused an alteration in the electrostatic surface potential of the pore helix. CONCLUSION: We propose two possible means by which the Tyr270His mutation causes hearing loss: a positively charged His270 side chain might enhance the helix dipole moment of the pore helix, thereby destabilizing the helix and/or the pore region, or it might disturb transport of K+ through the channel by electrostatic repulsion.


Subject(s)
Hearing Loss, Sensorineural/genetics , KCNQ Potassium Channels/chemistry , KCNQ Potassium Channels/genetics , Models, Molecular , Mutation, Missense , Amino Acid Sequence , Amino Acid Substitution , Audiometry , Base Sequence , DNA Mutational Analysis , Hearing Loss, Sensorineural/physiopathology , Humans , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Static Electricity
19.
J Comp Neurol ; 520(8): 1702-20, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22121026

ABSTRACT

The guidance of axonal projections to ipsilateral and contralateral regions is essential for integration of bilateral sensory information and coordination of movement. In the development of olivocerebellar projections, newborn neurons of inferior olivary (IO) nuclei ventrally migrate from the hindbrain rhombic lip to the floor plate (FP). The cell bodies of IO neurons cannot cross the FP but their axons can, and thus IO neurons project their axons only to the contralateral cerebellar cortex. The molecular mechanisms determining the contralateral axonal projections of IO neurons, however, are obscure. The IO neurons and their axons express EphA4, whereas the FP expresses an EphA4 ligand, EphrinB3, from embryonic day 12.5. Therefore, we tested whether EphA4-deficient mice (EphA4(-/-) ) would show impairment in the development of olivocerebellar projections. We found that, in EphA4(-/-) embryos, some of the IO neurons projected their axons to the ipsilateral cerebellar cortex because the cell bodies of the IO neurons abnormally crossed the FP. Furthermore, even in adults, EphA4(-/-) cerebella were bilaterally innervated by unilateral IO subnuclei. These observations indicate that EphA4 is involved in the contralateral axonal projections of IO neurons by preventing their cell bodies from crossing the midline FP.


Subject(s)
Cell Movement/physiology , Functional Laterality/physiology , Neurogenesis/physiology , Neurons/cytology , Olivary Nucleus/embryology , Receptor, EphA4/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Body Patterning/physiology , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred ICR , Mice, Knockout , Neurons/metabolism , Olivary Nucleus/growth & development , Receptor, EphA4/deficiency
20.
J Comp Neurol ; 519(13): 2594-614, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21456012

ABSTRACT

The adult cerebellum is organized into longitudinal compartments that are revealed by specific axonal projections (olivocerebellar and corticonuclear projections). These compartments in the adult cerebellum are closely correlated with the striped expression of zebrin II (aldolase C), a late-onset marker of Purkinje cells. Similarly, the embryonic cerebellum is organized into longitudinal compartments that are revealed by striped expression of other genes (early-onset markers). The cerebellar compartments are thought to be the basic and functional subdivisions of the cerebellum. However, the relationship between the embryonic (early-onset) and the adult (late-onset) compartments has remained unknown, because the pattern of the embryonic compartments is distinct from that of the adult compartments. To examine this issue, we labeled Purkinje cells (PCs) born at embryonic day (E) 10.5, E11.5, and E12.5 by using an adenoviral vector and traced their fated positions in the adult cerebellum. By comparing the striped distribution of each cohort of birth date-related PCs with the striped pattern of zebrin II immunoreactivity (zebrin II bands) in the entire adult cerebellum, we found that the striped distribution of PCs correlated strikingly with zebrin II bands. Generally, a single early-onset compartment was transformed directly into a single late-onset compartment. Therefore, our observation also indicated the close correlation between the compartments formed by birth date-related PCs and olivocerebellar projections. Furthermore, we found that the cerebellum was composed of three units showing lateral-to-medial developmental gradients, as revealed by the birth dates of PCs. The results suggest that PC birth dates play an important role in organizing cerebellar compartmentalization.


Subject(s)
Cerebellum/anatomy & histology , Cerebellum/embryology , Purkinje Cells/physiology , Adenoviridae , Animals , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Mice , Nerve Tissue Proteins/metabolism , Pregnancy , Purkinje Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...