Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
Auris Nasus Larynx ; 51(1): 82-85, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37573175

RESUMEN

Mitochondrial DNA mutations such as A3243G or A1555G are widely reported to cause hearing loss, but few reports exist on the A8296G mutation, which can also cause hearing loss. This report presents the case of a patient with the A8296G mutation and severe bilateral sensorineural hearing loss (SNHL) that progressed over two decades. The patient had no history of diabetes, but did have a family history of SNHL in her father and maternal grandmother. She was first diagnosed with SNHL at 45 years of age, and an A8296G mutation was found. The hearing threshold in the low-frequency range of the right ear was preserved at diagnosis, but eventually declined resulting in severe bilateral hearing loss by the age of 66 years, and cochlear implantation (CI) was performed in the left ear. The hearing threshold three months after CI was 25-45 dB HL, and the phoneme speech discrimination score in the left ear improved from 20% without CI to 74% with CI. SNHL patients with the A8295G mutation are good candidates for treatment with CI.


Asunto(s)
Implantación Coclear , Implantes Cocleares , Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Percepción del Habla , Humanos , Femenino , Anciano , Implantación Coclear/métodos , ADN Mitocondrial/genética , Pérdida Auditiva/complicaciones , Pérdida Auditiva Sensorineural/cirugía , Pérdida Auditiva Bilateral/genética , Pérdida Auditiva Bilateral/complicaciones , Mutación , Implantes Cocleares/efectos adversos
2.
Otol Neurotol ; 44(4): 360-366, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36804529

RESUMEN

OBJECTIVE: To investigate the diagnostic yield of targeted next-generation sequencing using hearing loss panels and to identify patient-related factors that are associated with a definite genetic cause. STUDY DESIGN: Retrospective chart review. SETTING: Tertiary referral center. PATIENTS: Children with congenital or late-onset, bilateral sensorineural hearing loss. INTERVENTIONS: Diagnostic. MAIN OUTCOME MEASURES: The number of patients with a definite genetic diagnosis. RESULTS: We report on 238 patients with hearing loss: 130 were male and 108 were female. About 55% had congenital hearing loss. A genetic cause was identified in 94 of the patients (39.5%), with 72.3% of these showing nonsyndromic and 27.6% showing syndromic hearing loss. The diagnostic yield was highest among North African patients (66.7%). A multiple linear regression model shows that profound hearing loss, family history of hearing loss, congenital hearing loss, and North African ethnicity are significantly related to identifying a genetic cause. CONCLUSIONS: Targeted next-generation sequencing using a panel of hearing loss genes identified a genetic diagnosis in almost 40% of children with bilateral sensorineural hearing loss. We describe the predictors of a genetic diagnosis, and this information may be used during genetic counseling.


Asunto(s)
Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Humanos , Niño , Masculino , Femenino , Estudios Retrospectivos , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Sordera/complicaciones , Pérdida Auditiva Bilateral/diagnóstico , Pérdida Auditiva Bilateral/genética , Pérdida Auditiva/complicaciones , Secuenciación de Nucleótidos de Alto Rendimiento
3.
JAMA Otolaryngol Head Neck Surg ; 149(3): 212-222, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36633841

RESUMEN

Importance: In the US, most childhood-onset bilateral sensorineural hearing loss is genetic, with more than 120 genes and thousands of different alleles known. Primary treatments are hearing aids and cochlear implants. Genetic diagnosis can inform progression of hearing loss, indicate potential syndromic features, and suggest best timing for individualized treatment. Objective: To identify the genetic causes of childhood-onset hearing loss and characterize severity, progression, and cochlear implant success associated with genotype in a single large clinical cohort. Design, Setting, and Participants: This cross-sectional analysis (genomics) and retrospective cohort analysis (audiological measures) were conducted from 2019 to 2022 at the otolaryngology and audiology clinics of Seattle Children's Hospital and the University of Washington and included 449 children from 406 families with bilateral sensorineural hearing loss with an onset younger than 18 years. Data were analyzed between January and June 2022. Main Outcomes and Measures: Genetic diagnoses based on genomic sequencing and structural variant analysis of the DNA of participants; severity and progression of hearing loss as measured by audiologic testing; and cochlear implant success as measured by pediatric and adult speech perception tests. Hearing thresholds and speech perception scores were evaluated with respect to age at implant, months since implant, and genotype using a multivariate analysis of variance and covariance. Results: Of 406 participants, 208 (51%) were female, 17 (4%) were African/African American, 32 (8%) were East Asian, 219 (54%) were European, 53 (13%) were Latino/Admixed American, and 16 (4%) were South Asian. Genomic analysis yielded genetic diagnoses for 210 of 406 families (52%), including 55 of 82 multiplex families (67%) and 155 of 324 singleton families (48%). Rates of genetic diagnosis were similar for children of all ancestries. Causal variants occurred in 43 different genes, with each child (with 1 exception) having causative variant(s) in only 1 gene. Hearing loss severity, affected frequencies, and progression varied by gene and, for some genes, by genotype within gene. For children with causative mutations in MYO6, OTOA, SLC26A4, TMPRSS3, or severe loss-of-function variants in GJB2, hearing loss was progressive, with losses of more than 10 dB per decade. For all children with cochlear implants, outcomes of adult speech perception tests were greater than preimplanted levels. Yet the degree of success varied substantially by genotype. Adjusting for age at implant and interval since implant, speech perception was highest for children with hearing loss due to MITF or TMPRSS3. Conclusions and Relevance: The results of this cross-sectional study suggest that genetic diagnosis is now sufficiently advanced to enable its integration into precision medical care for childhood-onset hearing loss.


Asunto(s)
Implantación Coclear , Implantes Cocleares , Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Percepción del Habla , Adulto , Femenino , Niño , Humanos , Masculino , Estudios Transversales , Estudios Retrospectivos , Sordera/cirugía , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/cirugía , Pérdida Auditiva Bilateral/diagnóstico , Pérdida Auditiva Bilateral/genética , Proteínas de la Membrana , Proteínas de Neoplasias , Serina Endopeptidasas
4.
Am J Med Genet A ; 185(3): 937-944, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33438842

RESUMEN

Townes-Brocks syndrome (TBS) is a rare autosomal dominant syndrome, resulting from heterozygous variant in SALL1 gene and initially characterized by the triad of anorectal, thumb, and ear malformations. Essentially described in children, adult case reports are uncommon. Renal involvement has already been reported in adults and children but poorly described. Structural abnormalities such as hypodysplasia, unilateral renal agenesis or multicystic kidneys have been described, as well as functional impairment (with or without structural abnormalities) that may progress to end-stage renal disease (ESRD). We report two adult cases (mother and daughter) which exhibited kidney hypoplasia (focal and segmental glomerulosclerosis for the mother) and ESRD. The mother had unilateral polydactyly. TBS was suggested after physical examination. TBS diagnosis was confirmed by identification of a SALL1 variant. We conducted a literature review to evaluate the renal anomalies in TBS cases diagnosed in adulthood. Among 44 adult cases of TBS with genetic confirmation (including our two cases), 10 had kidney disease. The circumstances of renal failure diagnosis were incidental findings (2/5), gout (2/5), or repeated episodes of pyelonephritis (1/5). The median age of kidney disease diagnosis was 30 years old and of renal transplant 49 years old. The most frequent renal malformation was bilateral kidney hypoplasia. TBS is probably underestimated in adulthood and this report highlights that less obvious elements of morphology such as dysplasic ears can facilitate the diagnosis of TBS. As long-term prognosis of renal involvement in TBS patients remains largely unknown, a regular evaluation is required throughout life for patients.


Asunto(s)
Ano Imperforado/complicaciones , Pérdida Auditiva Sensorineural/complicaciones , Fallo Renal Crónico/etiología , Pulgar/anomalías , Factores de Transcripción/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Aborto Habitual/genética , Ano Imperforado/diagnóstico , Ano Imperforado/genética , Diagnóstico Tardío , Oído Externo/anomalías , Femenino , Síndrome del Dedo del Pie en Martillo/genética , Pérdida Auditiva Bilateral/genética , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Humanos , Fallo Renal Crónico/cirugía , Fallo Renal Crónico/terapia , Trasplante de Riñón , Persona de Mediana Edad , Linaje , Enfermedades del Sistema Nervioso Periférico/genética , Fenotipo , Polidactilia/genética , Diálisis Renal , Distrofias Retinianas/genética
5.
Ear Nose Throat J ; 100(3_suppl): 333S-336S, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32791904

RESUMEN

We report a 6-year-old girl with progressive bilateral conductive hearing loss for 2 years. She passed the newborn hearing screening conducted with otoacoustic emissions testing and had a normal development of speech and language, which indicated that her deafness was delayed-onset. She also had congenital proximal interphalangeal joints. Proximal symphalangism was confirmed by genetic testing (NOG gene: c.406C > T, p.R136C). Bilateral stapes ankyloses were proved by surgery and her hearing was improved after stapedotomy by over 30 dB. Besides, this case should remind clinicians to carefully distinguish NOG gene-related deafness from congenital ossicular malformation and pediatric otosclerosis.


Asunto(s)
Proteínas Portadoras/genética , Articulaciones de los Dedos/anomalías , Pérdida Auditiva Bilateral/diagnóstico , Pérdida Auditiva Conductiva/diagnóstico , Artropatías/congénito , Estribo/anomalías , Audiometría de Tonos Puros , Niño , Diagnóstico Diferencial , Femenino , Pérdida Auditiva Bilateral/genética , Pérdida Auditiva Conductiva/genética , Humanos , Artropatías/diagnóstico , Artropatías/genética , Ilustración Médica , Cirugía del Estribo
6.
Genes (Basel) ; 11(12)2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33333757

RESUMEN

BACKGROUND: We have investigated the main genetic causes for non-syndromic hearing impairment (NSHI) in the hearing impairment individuals from the North-Eastern Romania and proposed a cost-effective diagnosis protocol. METHODS: MLPA followed by Sanger Sequencing were used for all 291 patients included in this study. RESULTS: MLPA revealed abnormal results in 141 cases (48.45%): 57 (40.5%) were c.35delG homozygous, 26 (18.44%) were c.35delG heterozygous, 14 (9.93%) were compound heterozygous and 16 (11.35%) had other types of variants. The entire coding region of GJB2 was sequenced and out of 150 patients with normal results at MLPA, 29.33% had abnormal results: variants in heterozygous state: c.71G>A (28%), c.457G>A (20%), c.269T>C (12%), c.109G>A (12%), c.100A>T (12%), c.551G>C (8%). Out of 26 patients with c.35delG in heterozygous state, 38.46% were in fact compound heterozygous. CONCLUSIONS: We identified two variants: c.109G>A and c.100A>T that have not been reported in any study from Romania. MLPA is an inexpensive, rapid and reliable technique that could be a cost-effective diagnosis method, useful for patients with hearing impairment. It can be adaptable for the mutation spectrum in every population and followed by Sanger sequencing can provide a genetic diagnosis for patients with different degrees of hearing impairment.


Asunto(s)
Pérdida Auditiva/genética , Reacción en Cadena de la Polimerasa Multiplex , Adolescente , Adulto , Audiometría/métodos , Niño , Preescolar , Conexina 26/genética , Análisis Costo-Beneficio , Femenino , Estudios de Asociación Genética , Genotipo , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/economía , Pérdida Auditiva/epidemiología , Pérdida Auditiva Bilateral/genética , Humanos , Lactante , Masculino , Tamizaje Masivo , Proteínas de la Membrana/genética , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa Multiplex/economía , Mutación Puntual , Rumanía/epidemiología , Análisis de Secuencia de ADN , Adulto Joven
7.
Mitochondrion ; 54: 57-64, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32659360

RESUMEN

Pathogenic point mutations of mitochondrial DNA (mtDNA) are associated with a large number of heterogeneous diseases involving multiple systems with which patients may present with a wide range of clinical phenotypes. In this study, we describe a novel heteroplasmic missense mutation, m.11406 T > A, of the ND4 gene encoding the subunit 4 of mitochondrial complex I in a 32-year-old woman with recurrent epileptic seizure, headache and bilateral hearing loss. Skeletal muscle histochemistry demonstrated that approximately 20% of fibers were cytochrome C oxidase (COX) deficient with increased activity of succinate dehydrogenase (SDH). Further investigations in muscle specimens showed significantly reduced level of ND4 protein. It is interesting that the subunits of complex I (ND1 and NDFUB8) and complex IV(CO1) were also remarkably decreased. These findings indicate that ND1, NDFUB8 and CO1 are more susceptible than other subunits to mutations in the mitochondrial ND4 gene.


Asunto(s)
Pérdida Auditiva Bilateral/etiología , Síndrome MELAS/diagnóstico por imagen , Mutación Missense , NADH Deshidrogenasa/genética , Convulsiones/etiología , Adulto , Femenino , Predisposición Genética a la Enfermedad , Pérdida Auditiva Bilateral/genética , Humanos , Síndrome MELAS/genética , Imagen por Resonancia Magnética , Masculino , Modelos Moleculares , NADH Deshidrogenasa/química , Linaje , Polimorfismo de Nucleótido Simple , Convulsiones/genética
8.
Am J Audiol ; 29(2): 236-243, 2020 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-32437266

RESUMEN

Objectives The purpose of this study is to ascertain the etiology of bilateral sensorineural hearing loss (SNHL) in children aged ≤ 18 years living in Shandong province. Method Data were taken from a cross-sectional study, which was conducted between 2015 and 2017. The study included children aged ≤ 18 years, recruited from special schools for children with hearing loss and from hearing rehabilitation centers in Shandong province of China. Children were screened for bilateral SNHL through audiological testing. Clinical examination, genetic testing, and structured interviews were conducted for those children who were identified as having hearing loss to identify the potential cause. Results The etiology of bilateral SNHL in our sample was genetic in 874 (39.3%), acquired in 650 (29.3%), and unknown in 697 (31.4%) children. Among children with acquired SNHL, the cause was maternal viral infection in 75 (11.5%); perinatal factors in 238 (36.6%); meningitis, measles, and mumps in 146 (22.5%); and ototoxic exposure in 117 (18%) children. Among the children with genetic SNHL, only 44 (4.9%) were identified as having syndromic hearing loss, and the remainder (95.1%) were classified as nonsyndromic hearing loss. Conclusion The findings indicated that nearly 30% of bilateral SNHL in Shandong province could be preventable through immunization, early prenatal diagnosis, proper treatment of infections, and avoidance of prescription of ototoxic drugs. This finding emphasizes the need for programs aimed at improving the health services at primary and secondary levels of health care, which will in turn prevent childhood hearing loss.


Asunto(s)
Pérdida Auditiva Bilateral/etiología , Pérdida Auditiva Sensorineural/etiología , Adolescente , Antibacterianos/efectos adversos , Asfixia Neonatal/complicaciones , Audiometría , Niño , Preescolar , China , Conexina 26/genética , Estudios Transversales , ADN Mitocondrial/genética , Síndrome de Down/complicaciones , Femenino , Gentamicinas/efectos adversos , Síndrome de Goldenhar/complicaciones , Pérdida Auditiva Bilateral/inducido químicamente , Pérdida Auditiva Bilateral/genética , Pérdida Auditiva Sensorineural/inducido químicamente , Pérdida Auditiva Sensorineural/genética , Infecciones por Herpesviridae/complicaciones , Humanos , Hiperbilirrubinemia/complicaciones , Hipertensión Inducida en el Embarazo , Lactante , Recién Nacido de Bajo Peso , Recien Nacido Prematuro , Kanamicina/efectos adversos , Masculino , Sarampión/complicaciones , Síndrome de Meige/complicaciones , Meningitis/complicaciones , Síndrome de Mobius/complicaciones , Paperas/complicaciones , Ototoxicidad , Neumonía/complicaciones , Embarazo , Complicaciones Infecciosas del Embarazo , ARN Ribosómico/genética , Síndrome de Rubéola Congénita/complicaciones , Transportadores de Sulfato/genética , Virosis/complicaciones , Virosis/congénito , Síndrome de Waardenburg/complicaciones
9.
Biochim Biophys Acta Mol Basis Dis ; 1866(6): 165742, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32105826

RESUMEN

Transforming growth factor beta-activated kinase 1 (TAK1) is a highly conserved kinase protein encoded by MAP3K7, and activated by multiple extracellular stimuli, growth factors and cytokines. Heterozygous variants in MAP3K7 cause the cardiospondylocarpofacial syndrome (CSCFS) which is characterized by short stature, dysmorphic facial features, cardiac septal defects with valve dysplasia, and skeletal anomalies. CSCFS has been described in seven patients to date and its molecular pathogenesis is only partially understood. Here, the functional effects of the MAP3K7 c.737-7A > G variant, previously identified in a girl with CSCFS and additional soft connective tissue features, were explored. This splice variant generates an in-frame insertion of 2 amino acid residues in the kinase domain of TAK1. Computational analysis revealed that this in-frame insertion alters protein dynamics in the kinase activation loop responsible for TAK1 autophosphorylation after binding with its interactor TAB1. Co-immunoprecipitation studies demonstrate that the ectopic expression of TAK1-mutated protein impairs its ability to physically bind TAB1. In patient's fibroblasts, MAP3K7 c.737-7A > G variant results in reduced TAK1 autophosphorylation and dysregulation of the downstream TAK1-dependent signaling pathway. TAK1 loss-of-function is associated with an impaired TGFß-mediated α-SMA cytoskeleton assembly and cell migration, and defective autophagy process. These findings contribute to our understanding of the molecular pathogenesis of CSCFS and might offer the rationale for the design of novel therapeutic targets.


Asunto(s)
Anomalías Múltiples/genética , Actinas/genética , Autofagia/genética , Pérdida Auditiva Bilateral/genética , Quinasas Quinasa Quinasa PAM/genética , Insuficiencia de la Válvula Mitral/genética , Osteosclerosis/genética , Anomalías Múltiples/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/genética , Niño , Citoesqueleto/genética , Femenino , Fibroblastos/metabolismo , Pérdida Auditiva Bilateral/fisiopatología , Humanos , Mutación con Pérdida de Función/genética , Insuficiencia de la Válvula Mitral/fisiopatología , Mutación/genética , Osteosclerosis/fisiopatología , Fosforilación/genética , Polimorfismo de Nucleótido Simple/genética , Unión Proteica/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética
10.
Mol Genet Genomic Med ; 7(12): e995, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31595699

RESUMEN

BACKGROUND: Childhood hearing impairment affects language and cognitive development. Profound congenital sensorineural hearing impairment can be due to an abnormal cochleovestibular nerve (CVN) and cochleovestibular malformations, however, the etiology of these conditions remains unclear. METHODS: We used a trio-based exome sequencing approach to unravel the underlying molecular etiology of a child with a rare nonsyndromic CVN abnormality and cochlear hypoplasia. Clinical and imaging data were also reviewed. RESULTS: We identified a de novo missense variant [p(Asn174Tyr)] in the DNA-binding Homeodomain of SIX1, a gene which previously has been associated with autosomal dominant hearing loss (ADHL) and branchio-oto-renal or Branchio-otic syndrome, a condition not seen in this patient. CONCLUSIONS: SIX1 has an important function in otic vesicle patterning during embryogenesis, and mice show several abnormalities to their inner ear including loss of inner ear innervation. Previous reports on patients with SIX1 variants lack imaging data and nonsyndromic AD cases were reported to have no inner ear malformations. In conclusion, we show that a de novo variant in SIX1 in a patient with sensorineural hearing loss leads to cochleovestibular malformations and abnormalities of the CVN, without any other abnormalities. Without proper interventions, severe to profound hearing loss is devastating to both education and social integration. Choosing the correct intervention can be challenging and a molecular diagnosis may adjust intervention and improve outcomes, especially for rare cases.


Asunto(s)
Cóclea/anomalías , Pérdida Auditiva Bilateral/genética , Pérdida Auditiva Sensorineural/genética , Proteínas de Homeodominio/genética , Mutación Missense , Nervio Vestibulococlear/anomalías , Niño , Proteínas de Homeodominio/química , Humanos , Masculino , Linaje , Dominios Proteicos , Secuenciación del Exoma
11.
Int J Pediatr Otorhinolaryngol ; 125: 92-97, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31276892

RESUMEN

OBJECTIVES: The aim of the study was to clarify differences in the prevalence and features of bony malformations in inner ear between congenital unilateral sensorineural hearing loss (USNHL) and congenital bilateral sensorineural hearing loss (BSNHL). METHODS: We conducted a retrospective study of 378 consecutive infants referred from routine newborn hearing screening in the past 18 years. Clinical background, audiological data, and temporal bone computed tomography (CT) findings were analyzed. The prevalence of malformations between USNHL and BSNHL groups were compared using the Chi-square test. RESULTS: The proportion of family history of hearing loss was significantly higher in infants with BSNHL than in those with USNHL (26/107 [24.3%] vs. 4/105 [3.7%]; p = 0.0001). Temporal bone CT scanning revealed significantly a higher prevalence of inner ear malformations in infants with USNHL than in those with BSNHL (93/109 [85.3%] vs. 4/107 [3.7%]; p < 0.0001). The most frequent anomaly in USNHL was cochlear nerve canal stenosis (69.7%), followed by cochlear malformations (20.2%), and narrow internal auditory canal (17.4%). Four infants with BSNHL accompanied by inner ear anomaly had complications such as Down's syndrome, developmental delay, or epilepsy. CONCLUSIONS: The prevalence of bony malformations in inner ear and/or IAC was markedly higher in infants with congenital USNHL than in infants with BSNHL. Temporal bone CT scanning may help to clarify the etiology of congenital hearing loss, especially in USNHL.


Asunto(s)
Oído Interno/anomalías , Pérdida Auditiva Bilateral/congénito , Pérdida Auditiva Sensorineural/congénito , Pérdida Auditiva Unilateral/congénito , Oído Interno/diagnóstico por imagen , Femenino , Predisposición Genética a la Enfermedad , Pérdida Auditiva Bilateral/genética , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Unilateral/genética , Humanos , Lactante , Recién Nacido , Masculino , Prevalencia , Estudios Retrospectivos , Hueso Temporal/anomalías , Hueso Temporal/diagnóstico por imagen , Tomografía Computarizada por Rayos X
12.
PLoS One ; 14(4): e0215212, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30973918

RESUMEN

Hearing loss (HL) is a common sensory disorder. More than half of HL cases can be attributed to genetic causes. There is no effective therapy for genetic HL at present, early diagnosis to reduce the incidence of genetic HL is important for clinical intervention in genetic HL. Previous studies have identified 111 nonsyndromic hearing loss genes. The most frequently mutated genes identified in NSHL patients in China include GJB2, SLC26A4, and the mitochondrial gene MT-RNR1. It is important to develop HL gene panels in Chinese population, which allow for etiologic diagnosis of both SHL and NSHL. In this study, a total of 220 unrelated Han Chinese patients with bilateral progressive SNHL and 50 unrelated healthy controls were performed Single nucleotide polymorphism (SNP) genotyping using an improved multiplex ligation detection reaction (iMLDR) technique, is to simultaneously detect a total of 32 mutations in ten HL genes, covering all currently characterized mutations involved in the etiology of nonsyndromic or syndromic hearing loss in the Chinese population. The 49 positive samples with known mutations were successfully detected using the iMLDR Technique. For 171 SNHL patients, gene variants were found in 57 cases (33.33%), among which, 30 patients carried mutations in GJB2, 14 patients carried mutations in SLC26A4, seven patients carried mutations in GJB3, and six patients carried mutations in MT-RNR1. The molecular etiology of deafness was confirmed in 12.9% (22/171) of patients carried homozygous variants. These results were verified by Sanger sequencing, indicating that the sensitivity and specificity of the iMLDR technique was 100%. We believe that the implementation of this population-specific technology at an efficient clinical level would have great value in HL diagnosis and treatment.


Asunto(s)
Pérdida Auditiva Bilateral/genética , Reacción en Cadena de la Polimerasa Multiplex/métodos , Mutación , Pueblo Asiatico/genética , Estudios de Casos y Controles , China , Conexina 26 , Conexinas/genética , Análisis Mutacional de ADN/métodos , Sordera/genética , Genes Mitocondriales , Humanos , Polimorfismo de Nucleótido Simple , Transportadores de Sulfato/genética
13.
Hum Mutat ; 40(2): 217-229, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30431684

RESUMEN

Ichthyosis follicularis, a distinct cutaneous entity reported in combination with atrichia, and photophobia has been associated with mutations in MBTPS2. We sought the genetic cause of a novel syndrome of ichthyosis follicularis, bilateral severe sensorineural hearing loss and punctate palmoplantar keratoderma in two families. We performed whole exome sequencing on three patients from two families. The pathogenicity and consequences of mutations were studied in the Xenopus oocyte expression system and by molecular modeling analysis. Compound heterozygous mutations in the GJB2 gene were discovered: a pathogenic c.526A>G; p.Asn176Asp, and a common frameshift mutation, c.35delG; p.Gly12Valfs*2. The p.Asn176Asp missense mutation was demonstrated to significantly reduce the cell-cell gap junction channel activity and increase the nonjunctional hemichannel activity in the Xenopus oocyte expression system. Molecular modeling analyses of the mutant Cx26 protein revealed significant changes in the structural characteristics and electrostatic potential of the Cx26, either in hemichannel or gap junction conformation. Thus, association of a new syndrome of an autosomal recessive disorder of ichthyosis follicularis, bilateral severe sensorineural hearing loss and punctate palmoplantar keratoderma with mutations in GJB2, expands the phenotypic spectrum of the GJB2-associated disorders. The findings attest to the complexity of the clinical consequences of different mutations in GJB2.


Asunto(s)
Conexinas/genética , Pérdida Auditiva Sensorineural/genética , Ictiosis/genética , Queratodermia Palmoplantar/genética , Animales , Conexina 26 , Pérdida Auditiva Bilateral/genética , Pérdida Auditiva Bilateral/patología , Pérdida Auditiva Sensorineural/patología , Humanos , Ictiosis/patología , Metaloendopeptidasas/genética , Mutación Missense/genética , Oocitos/crecimiento & desarrollo , Linaje , Piel/metabolismo , Xenopus/genética
14.
Gene ; 652: 59-65, 2018 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-29408584

RESUMEN

Mitochondrial DNA mutations play an important role in hereditary hearing loss. The present study aimed at identifying more novel genetic variants of mitochondrial DNA. Complete Mitochondrial genomes were detected in 97 Northwestern Chinese probands with non-syndromic hearing loss and 376 control subjects. The variants were evaluated for the pathogenicity using the following criteria: (1) present in <1% of the controls, (2) evolutional conservation, (3) potential structural and functional alterations. Mutational analysis in probands identified 706 variants, including 180 variants in the D loop region, 27 variants in the 12S rRNA gene, 29 variants in the 16S rRNA gene, 122 missense variants and 312 silent variants in the protein coding gene, 29 variants in the tRNA genes and 7 variants in the non-coding region. After further analysis assessed for the potential structural and functional significance, we identified 5 new candidate variants for hearing loss: 12S rRNA1473C>T, tRNAPhe 614A>C, tRNALys 8339A>G, ND1 3866T>C and non-coding 5656A>G. Our findings may provide the role of these genes in hearing loss development in China and valuable information for the further understanding of pathogenic mechanism of hearing loss.


Asunto(s)
ADN Mitocondrial/genética , Genes Mitocondriales , Genoma Mitocondrial , Pérdida Auditiva Bilateral/genética , ARN Ribosómico/genética , Adolescente , Adulto , Pueblo Asiatico , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Pérdida Auditiva Bilateral/diagnóstico , Pérdida Auditiva Bilateral/etnología , Pérdida Auditiva Bilateral/fisiopatología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Mutación , Linaje , ARN Ribosómico 16S/genética , ARN de Transferencia/genética
15.
Cytogenet Genome Res ; 153(2): 66-72, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29248929

RESUMEN

Unbalanced translocations involving X and Y chromosomes are rare and associated with a contiguous gene syndrome. The clinical phenotype is heterogeneous including mainly short stature, chondrodysplasia punctata, ichthyosis, hypogonadism, and intellectual disability. Here, we report 2 brothers with peculiar gestalt, short stature, and hearing loss, who harbor an X/Y translocation. Physical examination, brainstem acoustic potential evaluation, bone age, hormonal assessment, and X-ray investigations were performed. Because of their dysmorphic features, karyotyping, FISH, and aCGH were carried out. The probands had short stature, hypertelorism, midface hypoplasia, sensorineural hearing loss, normal intelligence as well as slight radial and ulnar bowing with brachytelephalangy. R-banding identified a derivative X chromosome with an abnormally expanded short arm. The mother was detected as a carrier of the same aberrant X chromosome. aCGH disclosed a 3.1-Mb distal deletion of chromosome region Xp22.33pter. This interval encompasses several genes, especially the short stature homeobox (SHOX) and arylsulfatase (ARSE) genes. The final karyotype of the probands was: 46,Y,der(X),t(X;Y)(p22;q12).ish der(X)(DXYS129-,DXYS153-)mat.arr[hg19] Xp22.33(61091_2689408)×1mat,Xp22.33(2701273_3258404)×0mat,Yq11.222q12 (21412851_59310245)×2. Herein, we describe a Moroccan family with a maternally inherited X/Y translocation and discuss the genotype-phenotype correlations according to the deleted genes.


Asunto(s)
Anomalías Múltiples/genética , Arilsulfatasas/genética , Cromosomas Humanos X/genética , Cromosomas Humanos Y/genética , Pérdida Auditiva Bilateral/genética , Pérdida Auditiva Sensorineural/genética , Translocación Genética , Arilsulfatasas/deficiencia , Cromosomas Humanos X/ultraestructura , Cromosomas Humanos Y/ultraestructura , Consanguinidad , Femenino , Humanos , Hipertelorismo/genética , Recién Nacido , Cariotipificación , Masculino , Persona de Mediana Edad , Marruecos , Linaje , Fenotipo , Radio (Anatomía)/anomalías , Escoliosis/genética , Hermanos , Cúbito/anomalías , Adulto Joven
16.
BMC Med Genet ; 18(1): 151, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29258540

RESUMEN

BACKGROUND: Low-frequency nonsyndromic hearing loss (LF-NSHL) is a rare, inherited disorder. Here, we report a family with LF-NSHL in whom a missense mutation was found in the Wolfram syndrome 1 (WFS1) gene. CASE PRESENTATION: Family members underwent audiological and imaging evaluations, including pure tone audiometry and temporal bone computed tomography. Blood samples were collected from two affected and two unaffected subjects. To determine the genetic background of hearing loss in this family, genetic analysis was performed using whole-exome sequencing. Among 553 missense variants, c.2419A → C (p.Ser807Arg) in WFS1 remained after filtering and inspection of whole-exome sequencing data. This missense mutation segregated with affected status and demonstrated an alteration to an evolutionarily conserved amino acid residue. Audiological evaluation of the affected subjects revealed nonprogressive LF-NSHL, with early onset at 10 years of age, but not to a profound level. CONCLUSION: This is the second report to describe a pathological mutation in WFS1 among Korean patients and the second to describe the mutation in a different ethnic background. Given that the mutation was found in independent families, p.S807R possibly appears to be a "hot spot" in WFS1, which is associated with LF-NSHL.


Asunto(s)
Sordera/genética , Pérdida Auditiva Bilateral/genética , Proteínas de la Membrana/genética , Mutación Missense/genética , Adolescente , Pueblo Asiatico/genética , Audiometría , Femenino , Humanos , Masculino , Persona de Mediana Edad , República de Corea , Hueso Temporal/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Secuenciación del Exoma
17.
Cytogenet Genome Res ; 153(2): 81-85, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29258104

RESUMEN

Partial monosomy 21 results in a great variability of clinical features that may be associated with the size and location of the deletion. In this study, we report a 22-month-old girl who showed a 45,XX,add(12)(p13)dn,-21 karyotype. The final cytogenomic result was 45,XX,der(12)t(12;21)(p13;q22.11) dn,-21.arr[hg19] 21q11.2q22.11(14824453_33868129)×1 revealing a deletion from 21pter to 21q22.11. Clinical manifestation of the patient included hypertonia, a long philtrum, epicanthic folds, low-set ears, and café-au-lait macules - a phenotype considered as mild despite the relatively large size of the deletion compared to patients from the literature.


Asunto(s)
Anomalías Múltiples/genética , Manchas Café con Leche/genética , Deleción Cromosómica , Cromosomas Humanos Par 21/ultraestructura , Cara/anomalías , Hipertonía Muscular/genética , Cromosomas Humanos Par 21/genética , Discapacidades del Desarrollo/genética , Femenino , Pérdida Auditiva Bilateral/genética , Humanos , Recién Nacido , Cariotipificación , Fenotipo , Escoliosis/genética
18.
Cytogenet Genome Res ; 153(4): 175-180, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29518772

RESUMEN

Ring chromosome 13 is a rare genetic condition with an incidence of 1/58,000 in live births. Major clinical features of patients with ring chromosome 13 include growth and developmental retardation, microcephaly, facial dysmorphism, ambiguous genitalia, anal atresia, eye malformations, retinoblastoma, and hand, foot, and toe abnormalities. The severity of the phenotype depends on the amount of genetic material lost during ring chromosome formation. Here, we report 2 cases with ring chromosome 13 at either end of the phenotypic spectrum.


Asunto(s)
Anomalías Múltiples/genética , Agenesia del Cuerpo Calloso/genética , Trastornos de los Cromosomas/genética , Cardiopatías Congénitas/genética , Microcefalia/genética , Astigmatismo/genética , Trastornos de los Cromosomas/patología , Cromosomas Humanos Par 13/genética , Cromosomas Humanos Par 13/ultraestructura , Hibridación Genómica Comparativa , Resultado Fatal , Femenino , Retardo del Crecimiento Fetal/genética , Pérdida Auditiva Bilateral/genética , Pérdida Auditiva Sensorineural/genética , Humanos , Lactante , Recién Nacido , Trastornos del Desarrollo del Lenguaje/genética , Fenotipo , Polihidramnios/etiología , Embarazo , Cromosomas en Anillo , Análisis de Matrices Tisulares
20.
Otol Neurotol ; 38(2): 173-179, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27861301

RESUMEN

BACKGROUND: Heterozygous mutations in GJB2 (MIM: 121011) encoding the gap junction protein connexin 26 are overrepresented in patient groups suffering from nonsyndromic sensorineural hearing impairment (HI) implying the involvement of additional genetic factors. Mutations in SLC26A4 (MIM: 605646), encoding the protein pendrin can cause both Pendred syndrome and autosomal recessive, nonsyndromic HI locus 4 type sensorineural HI (MIM: 600791). OBJECTIVES: Aim of this study was to investigate the role of SLC26A4 coding mutations in a nonsyndromic hearing impairment (NSHI) patient group bearing heterozygous GJB2 35delG mutations. DESIGN: We analyzed the 20 coding exons of SLC26A4 in a group of patients (n = 15) bearing heterozygous 35delG mutations and exclusively suffering from congenital HI. RESULTS: In a case of bilateral congenital hearing loss we identified a rare, novel SLC26A4 exon 2 splice donor mutation (c.164+1delG) predicted to truncate pendrin in the first cytoplasmic domain, as a compound heterozygote with the pathogenic missense mutation c.1061T>C (p.354F>S; rs111033243). CONCLUSIONS: Screening for SLC26A4 mutations may identify the genetic causes of hearing loss in patients bearing heterozygous mutations in GJB2. HYPOTHESIS: SLC26A4 coding mutations are genetic causes for nonsyndromic HI in patients bearing heterozygous GJB2 35delG mutations.


Asunto(s)
Pérdida Auditiva Bilateral/genética , Pérdida Auditiva Sensorineural/genética , Proteínas de Transporte de Membrana/genética , Adulto , Niño , Preescolar , Conexina 26 , Conexinas/genética , Exones , Femenino , Heterocigoto , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación , Transportadores de Sulfato , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...