Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(26): e2204084119, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35727972

RESUMEN

Discovery of deafness genes and elucidating their functions have substantially contributed to our understanding of hearing physiology and its pathologies. Here we report on DNA variants in MINAR2, encoding membrane integral NOTCH2-associated receptor 2, in four families underlying autosomal recessive nonsyndromic deafness. Neurologic evaluation of affected individuals at ages ranging from 4 to 80 y old does not show additional abnormalities. MINAR2 is a recently annotated gene with limited functional understanding. We detected three MINAR2 variants, c.144G > A (p.Trp48*), c.412_419delCGGTTTTG (p.Arg138Valfs*10), and c.393G > T, in 13 individuals with congenital- or prelingual-onset severe-to-profound sensorineural hearing loss (HL). The c.393G > T variant is shown to disrupt a splice donor site. We show that Minar2 is expressed in the mouse inner ear, with the protein localizing mainly in the hair cells, spiral ganglia, the spiral limbus, and the stria vascularis. Mice with loss of function of the Minar2 protein (Minar2tm1b/tm1b) present with rapidly progressive sensorineural HL associated with a reduction in outer hair cell stereocilia in the shortest row and degeneration of hair cells at a later age. We conclude that MINAR2 is essential for hearing in humans and mice and its disruption leads to sensorineural HL. Progressive HL observed in mice and in some affected individuals and as well as relative preservation of hair cells provides an opportunity to interfere with HL using genetic therapies.


Asunto(s)
Pérdida Auditiva Sensorineural , Receptor Notch2 , Receptores de Superficie Celular , Animales , Pérdida Auditiva Sensorineural/genética , Humanos , Mutación con Pérdida de Función , Ratones , Receptor Notch2/genética , Receptor Notch2/metabolismo , Receptores de Superficie Celular/genética , Estereocilios/metabolismo
2.
Hum Genomics ; 17(1): 103, 2023 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-37996878

RESUMEN

BACKGROUND: We analyzed the genetic causes of sensorineural hearing loss in racial and ethnic minorities of South Florida by reviewing demographic, phenotypic, and genetic data on 136 patients presenting to the Hereditary Hearing Loss Clinic at the University of Miami. In our retrospective chart review, of these patients, half self-identified as Hispanic, and the self-identified racial distribution was 115 (86%) White, 15 (11%) Black, and 6 (4%) Asian. Our analysis helps to reduce the gap in understanding the prevalence, impact, and genetic factors related to hearing loss among diverse populations. RESULTS: The causative gene variant or variants were identified in 54 (40%) patients, with no significant difference in the molecular diagnostic rate between Hispanics and Non-Hispanics. However, the total solve rate based on race was 40%, 47%, and 17% in Whites, Blacks, and Asians, respectively. In Non-Hispanic Whites, 16 different variants were identified in 13 genes, with GJB2 (32%), MYO7A (11%), and SLC26A4 (11%) being the most frequently implicated genes. In White Hispanics, 34 variants were identified in 20 genes, with GJB2 (22%), MYO7A (7%), and STRC-CATSPER2 (7%) being the most common. In the Non-Hispanic Black cohort, the gene distribution was evenly dispersed, with 11 variants occurring in 7 genes, and no variant was identified in 3 Hispanic Black probands. For the Asian cohort, only one gene variant was found out of 6 patients. CONCLUSION: This study demonstrates that the diagnostic rate of genetic studies in hearing loss varies according to race in South Florida, with more heterogeneity in racial and ethnic minorities. Further studies to delineate deafness gene variants in underrepresented populations, such as African Americans/Blacks from Hispanic groups, are much needed to reduce racial and ethnic disparities in genetic diagnoses.


Asunto(s)
Pérdida Auditiva Sensorineural , Humanos , Asiático/genética , Negro o Afroamericano/genética , ADN/genética , Florida/epidemiología , Pérdida Auditiva Sensorineural/epidemiología , Pérdida Auditiva Sensorineural/genética , Hispánicos o Latinos/genética , Péptidos y Proteínas de Señalización Intercelular , Estudios Retrospectivos , Blanco/genética
3.
Am J Med Genet A ; 194(4): e63481, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37984424

RESUMEN

Chanarin-Dorfman syndrome is an autosomal recessively inherited disorder characterized by ichthyosis, sensorineural hearing loss, and hepatic dysfunction. We report on a 60-year-old female of Venezuelan descent who presented with congenital ichthyosis, progressive sensorineural hearing loss, and liver cirrhosis. We identify a heterozygous copy number deletion involving exon 1 and another heterozygous deletion involving exon 3 of the ABHD5 gene. Exon 2 is preserved. Both deletions were confirmed with RT-PCR. RNAseq from peripheral blood shows a reduction of ABHD5 expression overall and an absence of exon 3 expression, confirming the deleterious effects of the identified deletions. We present exonic deletions as a potentially common type of ABHD5 variation.


Asunto(s)
Pérdida Auditiva Sensorineural , Eritrodermia Ictiosiforme Congénita , Ictiosis , Errores Innatos del Metabolismo Lipídico , Enfermedades Musculares , Femenino , Humanos , Persona de Mediana Edad , Eritrodermia Ictiosiforme Congénita/complicaciones , Eritrodermia Ictiosiforme Congénita/diagnóstico , Eritrodermia Ictiosiforme Congénita/genética , Errores Innatos del Metabolismo Lipídico/genética , Enfermedades Musculares/genética , Ictiosis/complicaciones , Ictiosis/diagnóstico , Ictiosis/genética , Cirrosis Hepática , Pérdida Auditiva Sensorineural/complicaciones , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/genética
4.
Am J Med Genet A ; 194(6): e63563, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38352997

RESUMEN

Autosomal dominant sensorineural hearing loss (ADSNHL) is a genetically heterogeneous disorder caused by pathogenic variants in various genes, including MYH14. However, the interpretation of pathogenicity for MYH14 variants remains a challenge due to incomplete penetrance and the lack of functional studies and large families. In this study, we performed exome sequencing in six unrelated families with ADSNHL and identified five MYH14 variants, including three novel variants. Two of the novel variants, c.571G > C (p.Asp191His) and c.571G > A (p.Asp191Asn), were classified as likely pathogenic using ACMG and Hearing Loss Expert panel guidelines. In silico modeling demonstrated that these variants, along with p.Gly1794Arg, can alter protein stability and interactions among neighboring molecules. Our findings suggest that MYH14 causative variants may be more contributory and emphasize the importance of considering this gene in patients with nonsyndromic mainly post-lingual severe form of hearing loss. However, further functional studies are needed to confirm the pathogenicity of these variants.


Asunto(s)
Secuenciación del Exoma , Pérdida Auditiva Sensorineural , Cadenas Pesadas de Miosina , Miosina Tipo II , Linaje , Humanos , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Femenino , Masculino , Cadenas Pesadas de Miosina/genética , Adulto , Mutación/genética , Predisposición Genética a la Enfermedad , Niño , Genes Dominantes , Persona de Mediana Edad , Adolescente
5.
Am J Med Genet A ; 194(6): e63556, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38348595

RESUMEN

Phenotypic features of a hereditary connective tissue disorder, including craniofacial characteristics, hyperextensible skin, joint laxity, kyphoscoliosis, arachnodactyly, inguinal hernia, and diverticulosis associated with biallelic pathogenic variants in EFEMP1 have been previously described in four patients. Genome sequencing on a proband and her mother with comparable phenotypic features revealed that both patients were heterozygous for a stop-gain variant c.1084C>T (p.Arg362*). Complementary RNA-seq on fibroblasts revealed significantly reduced levels of mutant EFEMP1 transcript. Considering the absence of other molecular explanations, we extrapolated that EFEMP1 could be the cause of the patient's phenotypes. Furthermore, nonsense-mediated decay was demonstrated for the mutant allele as the principal mechanism for decreased levels of EFEMP1 mRNA. We provide strong clinical and genetic evidence for the haploinsufficiency of EFEMP1 due to nonsense-medicated decay to cause severe kyphoscoliosis, generalized hypermobility of joints, high and narrow arched palate, and potentially severe diverticulosis. To the best of our knowledge, this is the first report of an autosomal dominant EFEMP1-associated hereditary connective tissue disorder and therefore expands the phenotypic spectrum of EFEMP1 related disorders.


Asunto(s)
Enfermedades del Tejido Conjuntivo , Proteínas de la Matriz Extracelular , Haploinsuficiencia , Síndrome de Marfan , Fenotipo , Humanos , Haploinsuficiencia/genética , Femenino , Síndrome de Marfan/genética , Síndrome de Marfan/patología , Proteínas de la Matriz Extracelular/genética , Enfermedades del Tejido Conjuntivo/genética , Enfermedades del Tejido Conjuntivo/patología , Linaje , Mutación/genética , Degradación de ARNm Mediada por Codón sin Sentido/genética , Masculino , Adulto , Alelos , Predisposición Genética a la Enfermedad , Niño
6.
J Hum Genet ; 68(10): 657-669, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37217689

RESUMEN

Hearing loss (HL) is a common heterogeneous trait that involves variants in more than 200 genes. In this study, we utilized exome (ES) and genome sequencing (GS) to effectively identify the genetic cause of presumably non-syndromic HL in 322 families from South and West Asia and Latin America. Biallelic GJB2 variants were identified in 58 probands at the time of enrollment these probands were excluded. In addition, upon review of phenotypic findings, 38/322 probands were excluded based on syndromic findings at the time of ascertainment and no further evaluation was performed on those samples. We performed ES as a primary diagnostic tool on one or two affected individuals from 212/226 families. Via ES we detected a total of 78 variants in 30 genes and showed their co-segregation with HL in 71 affected families. Most of the variants were frameshift or missense and affected individuals were either homozygous or compound heterozygous in their respective families. We employed GS as a primary test on a subset of 14 families and a secondary tool on 22 families which were unsolved by ES. Although the cumulative detection rate of causal variants by ES and GS is 40% (89/226), GS alone has led to a molecular diagnosis in 7 of 14 families as the primary tool and 5 of 22 families as the secondary test. GS successfully identified variants present in deep intronic or complex regions not detectable by ES.


Asunto(s)
Sordera , Pérdida Auditiva , Humanos , Sordera/genética , Pérdida Auditiva/genética , Pérdida Auditiva/diagnóstico , Fenotipo , Homocigoto , Mutación , Linaje
7.
Am J Med Genet A ; 191(4): 1044-1049, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36628575

RESUMEN

Phenotypic features of KBG syndrome include craniofacial anomalies, short stature, cognitive disability and behavioral findings. The syndrome is caused by heterozygous pathogenic single nucleotide variants and indels in ANKRD11, or a heterozygous deletion of 16q24.3 that includes ANKRD11. We performed genome sequencing on a patient with clinical manifestations of KBG syndrome including distinct craniofacial features as well as a history of mild intellectual disability and attention-deficit hyperactivity disorder. This led to the identification of a 43 kb intragenic deletion of ANKRD11 affecting the first noncoding exon while leaving the coding regions intact. Review of the literature shows that this is the smallest 5' deletion affecting only the noncoding exons of ANKRD11. Real-time polymerase chain reaction demonstrated that the copy number variant was not present in either of the proband's parents, suggesting it occurred de novo. RNA expression analysis demonstrated significantly decreased transcript abundance compared to controls. This provides new evidence for haploinsufficiency as a mechanism of disease in KBG syndrome.


Asunto(s)
Anomalías Múltiples , Enfermedades del Desarrollo Óseo , Discapacidad Intelectual , Anomalías Dentarias , Humanos , Anomalías Múltiples/genética , Discapacidad Intelectual/genética , Enfermedades del Desarrollo Óseo/genética , Anomalías Dentarias/genética , Facies , Proteínas Represoras/genética , Deleción Cromosómica , Factores de Transcripción/genética , Fenotipo
8.
Am J Med Genet A ; 191(7): 1911-1916, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36987712

RESUMEN

Recurrent de novo missense variants in H4 histone genes have recently been associated with a novel neurodevelopmental syndrome that is characterized by intellectual disability and developmental delay as well as more variable findings that include short stature, microcephaly, and facial dysmorphisms. A 4-year-old male with autism, developmental delay, microcephaly, and a happy demeanor underwent evaluation through the Undiagnosed Disease Network. He was clinically suspected to have Angelman syndrome; however, molecular testing was negative. Genome sequencing identified the H4 histone gene variant H4C5 NM_003545.4: c.295T>C, p.Tyr99His, which parental testing confirmed to be de novo. The variant met criteria for a likely pathogenic classification and is one of the seven known disease-causing missense variants in H4C5. A comparison of our proband's findings to the initial description of the H4-associated neurodevelopmental syndrome demonstrates that his phenotype closely matches the spectrum of those reported among the 29 affected individuals. As such, this report corroborates the delineation of neurodevelopmental syndrome caused by de novo missense H4 gene variants. Moreover, it suggests that cases of clinically suspected Angelman syndrome without molecular confirmation should undergo exome or genome sequencing, as novel neurodevelopmental syndromes with phenotypes overlapping with Angelman continue to be discovered.


Asunto(s)
Síndrome de Angelman , Discapacidad Intelectual , Microcefalia , Trastornos del Neurodesarrollo , Masculino , Humanos , Síndrome de Angelman/diagnóstico , Síndrome de Angelman/genética , Microcefalia/genética , Histonas/genética , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Fenotipo , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/genética , Mutación Missense/genética
9.
Proc Natl Acad Sci U S A ; 116(4): 1347-1352, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30610177

RESUMEN

We have identified a GRAP variant (c.311A>T; p.Gln104Leu) cosegregating with autosomal recessive nonsyndromic deafness in two unrelated families. GRAP encodes a member of the highly conserved growth factor receptor-bound protein 2 (GRB2)/Sem-5/drk family of proteins, which are involved in Ras signaling; however, the function of the growth factor receptor-bound protein 2 (GRB2)-related adaptor protein (GRAP) in the auditory system is not known. Here, we show that, in mouse, Grap is expressed in the inner ear and the protein localizes to the neuronal fibers innervating cochlear and utricular auditory hair cells. Downstream of receptor kinase (drk), the Drosophila homolog of human GRAP, is expressed in Johnston's organ (JO), the fly hearing organ, and the loss of drk in JO causes scolopidium abnormalities. drk mutant flies present deficits in negative geotaxis behavior, which can be suppressed by human wild-type but not mutant GRAP. Furthermore, drk specifically colocalizes with synapsin at synapses, suggesting a potential role of such adaptor proteins in regulating actin cytoskeleton dynamics in the nervous system. Our findings establish a causative link between GRAP mutation and nonsyndromic deafness and suggest a function of GRAP/drk in hearing.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Adaptadora GRB2/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/metabolismo , Sordera/microbiología , Drosophila/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica/fisiología , Transducción de Señal/fisiología
10.
Hum Mol Genet ; 28(8): 1286-1297, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30561639

RESUMEN

Molecular mechanisms governing the development of the human cochlea remain largely unknown. Through genome sequencing, we identified a homozygous FOXF2 variant c.325A>T (p.I109F) in a child with profound sensorineural hearing loss (SNHL) associated with incomplete partition type I anomaly of the cochlea. This variant is not found in public databases or in over 1000 ethnicity-matched control individuals. I109 is a highly conserved residue in the forkhead box (Fox) domain of FOXF2, a member of the Fox protein family of transcription factors that regulate the expression of genes involved in embryogenic development as well as adult life. Our in vitro studies show that the half-life of mutant FOXF2 is reduced compared to that of wild type. Foxf2 is expressed in the cochlea of developing and adult mice. The mouse knockout of Foxf2 shows shortened and malformed cochleae, in addition to altered shape of hair cells with innervation and planar cell polarity defects. Expressions of Eya1 and Pax3, genes essential for cochlear development, are reduced in the cochleae of Foxf2 knockout mice. We conclude that FOXF2 plays a major role in cochlear development and its dysfunction leads to SNHL and developmental anomalies of the cochlea in humans and mice.


Asunto(s)
Cóclea/embriología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/fisiología , Adulto , Animales , Niño , Cóclea/metabolismo , Cóclea/fisiología , Desarrollo Embrionario , Femenino , Células Ciliadas Auditivas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Organogénesis , Factor de Transcripción PAX3/genética , Factor de Transcripción PAX3/fisiología , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal/genética , Secuenciación Completa del Genoma
11.
Am J Med Genet A ; 185(4): 1236-1241, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33427402

RESUMEN

Peroxisomes play an essential role in lipid metabolism via interaction with other intracellular organelles. The information about the role of the Acyl-CoA-binding domain containing-protein 5 (ACBD5) in these interactions in human cells is emerging. Moreover, a few patients with retinal dystrophy and leukodystrophy caused by pathogenic variants in ACBD5 have been recently introduced. Here, we present a 36-year-old female with retinal dystrophy, leukodystrophy, and psychomotor regression due to a novel homozygous variant in ACBD5. Our study adds to the growing knowledge of this peroxisomal disorder by providing phenotypic details of the first adult patient.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Predisposición Genética a la Enfermedad , Metabolismo de los Lípidos/genética , Proteínas de la Membrana/genética , Distrofias Retinianas/genética , Adulto , Femenino , Homocigoto , Humanos , Peroxisomas/genética , Peroxisomas/patología , Distrofias Retinianas/metabolismo , Distrofias Retinianas/patología
12.
J Inherit Metab Dis ; 43(6): 1199-1204, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32748411

RESUMEN

Galactosemia is a rare, treatable hereditary disorder of carbohydrate metabolism. We investigated the etiology of decreased GALT enzyme activity in a cohort of newborns referred by the Florida Newborn Screening Program with no detectable GALT variants in diagnostic molecular tests. Six affected individuals from four families with Guatemalan heritage were included. GALT enzyme activity ranged from 20% to 34% of normal. Clinical findings were unremarkable except for speech delay in two children. Via genome sequencing followed by Sanger confirmation we showed that all affected individuals were homozygous for a deep intronic GALT variant, c.1059+390A>G, which segregated as an autosomal recessive trait in all families. The intronic variant disrupts splicing and leads to a premature termination and is associated with a single haplotype flanking GALT, suggesting a founder effect. In conclusion, we present a deep intronic GALT variant leading to a biochemical variant form of galactosemia. This variant remains undiagnosed until it is specifically targeted in molecular testing.


Asunto(s)
Galactosemias/diagnóstico , Homocigoto , Mutación , UTP-Hexosa-1-Fosfato Uridililtransferasa/genética , Preescolar , Salud de la Familia , Femenino , Galactosemias/sangre , Galactosemias/genética , Pruebas Genéticas , Humanos , Lactante , Recién Nacido , Masculino , Tamizaje Neonatal , UTP-Hexosa-1-Fosfato Uridililtransferasa/deficiencia
13.
Hum Genet ; 138(10): 1071-1075, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31175426

RESUMEN

While the importance of tight junctions in hearing is well established, the role of Claudin- 9 (CLDN9), a tight junction protein, in human hearing and deafness has not been explored. Through whole-genome sequencing, we identified a one base pair deletion (c.86delT) in CLDN9 in a consanguineous family from Turkey with autosomal recessive nonsyndromic hearing loss. Three affected members of the family had sensorineural hearing loss (SNHL) ranging from moderate to profound in severity. The variant is predicted to cause a frameshift and produce a truncated protein (p.Leu29ArgfsTer4) in this single-exon gene. It is absent in public databases as well as in over 1000 Turkish individuals, and co-segregates with SNHL in the family. Our in vitro studies demonstrate that the mutant protein does not localize to cell membrane as demonstrated for the wild-type protein. Mice-lacking Cldn9 have been shown to develop SNHL. We conclude that CLDN9 is essential for proper audition in humans and its disruption leads to SNHL in humans.


Asunto(s)
Claudinas/genética , Sordera/diagnóstico , Sordera/genética , Genes Recesivos , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Claudinas/química , Claudinas/metabolismo , Biología Computacional/métodos , Análisis Mutacional de ADN , Femenino , Mutación del Sistema de Lectura , Humanos , Mutación , Linaje , Polimorfismo Genético , Transporte de Proteínas , Turquía , Secuenciación Completa del Genoma
14.
Clin Genet ; 96(6): 575-578, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31432506

RESUMEN

Auditory reception relies on the perception of mechanical stimuli by stereocilia and its conversion to electrochemical signal. Mechanosensory stereocilia are abundant in actin, which provides them with structural conformity necessary for perception of auditory stimuli. Out of three major classes of actin-bundling proteins, plastin 1 encoded by PLS1, is highly expressed in stereocilia and is necessary for their regular maintenance. A missense PLS1 variant associated with autosomal dominant hearing loss (HL) in a small family has recently been reported. Here, we present another PLS1 missense variant, c.805G > A (p.E269K), in a Turkish family with autosomal dominant non-syndromic HL confirming the causative role of PLS1 mutations in HL. We propose that HL due to the p.E269K variant is from the loss of a stable PLS1-ACTB interaction.


Asunto(s)
Genes Dominantes , Pérdida Auditiva/genética , Glicoproteínas de Membrana/genética , Proteínas de Microfilamentos/genética , Mutación/genética , Secuencia de Aminoácidos , Secuencia de Bases , Familia , Femenino , Humanos , Masculino , Glicoproteínas de Membrana/química , Proteínas de Microfilamentos/química , Proteínas Mutantes/química , Linaje , Turquía
15.
Proc Natl Acad Sci U S A ; 113(21): 5993-8, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27162350

RESUMEN

Hair cells of the inner ear, the mechanosensory receptors, convert sound waves into neural signals that are passed to the brain via the auditory nerve. Little is known about the molecular mechanisms that govern the development of hair cell-neuronal connections. We ascertained a family with autosomal recessive deafness associated with a common cavity inner ear malformation and auditory neuropathy. Via whole-exome sequencing, we identified a variant (c.2207G>C, p.R736T) in ROR1 (receptor tyrosine kinase-like orphan receptor 1), cosegregating with deafness in the family and absent in ethnicity-matched controls. ROR1 is a tyrosine kinase-like receptor localized at the plasma membrane. At the cellular level, the mutation prevents the protein from reaching the cellular membrane. In the presence of WNT5A, a known ROR1 ligand, the mutated ROR1 fails to activate NF-κB. Ror1 is expressed in the inner ear during development at embryonic and postnatal stages. We demonstrate that Ror1 mutant mice are severely deaf, with preserved otoacoustic emissions. Anatomically, mutant mice display malformed cochleae. Axons of spiral ganglion neurons show fasciculation defects. Type I neurons show impaired synapses with inner hair cells, and type II neurons display aberrant projections through the cochlear sensory epithelium. We conclude that Ror1 is crucial for spiral ganglion neurons to innervate auditory hair cells. Impairment of ROR1 function largely affects development of the inner ear and hearing in humans and mice.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Mutación , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Ganglio Espiral de la Cóclea/metabolismo , Animales , Axones/metabolismo , Axones/patología , Línea Celular , Células Ciliadas Auditivas/patología , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Humanos , Ratones , Ratones Mutantes , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Ganglio Espiral de la Cóclea/patología , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
16.
Hum Genet ; 137(6-7): 479-486, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29982980

RESUMEN

While recent studies have revealed a substantial portion of the genes underlying human hearing loss, the extensive genetic landscape has not been completely explored. Here, we report a loss-of-function variant (c.72delA) in MPZL2 in three unrelated multiplex families from Turkey and Iran with autosomal recessive nonsyndromic hearing loss. The variant co-segregates with moderate sensorineural hearing loss in all three families. We show a shared haplotype flanking the variant in our families implicating a single founder. While rare in other populations, the allele frequency of the variant is ~ 0.004 in Ashkenazi Jews, suggesting that it may be an important cause of moderate hearing loss in that population. We show that Mpzl2 is expressed in mouse inner ear, and the protein localizes in the auditory inner and outer hair cells, with an asymmetric subcellular localization. We thus present MPZL2 as a novel gene associated with sensorineural hearing loss.


Asunto(s)
Moléculas de Adhesión Celular/genética , Sordera/genética , Células Ciliadas Auditivas Internas/metabolismo , Pérdida Auditiva Sensorineural/genética , Animales , Sordera/fisiopatología , Oído Interno/crecimiento & desarrollo , Oído Interno/fisiopatología , Femenino , Frecuencia de los Genes , Genes Recesivos , Células Ciliadas Auditivas Internas/patología , Haplotipos/genética , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Irán/epidemiología , Judíos/genética , Masculino , Ratones , Mutación , Linaje , Células de Schwann/patología , Turquía
17.
Proc Natl Acad Sci U S A ; 111(27): 9864-8, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24958875

RESUMEN

In a large consanguineous Turkish kindred with recessive nonsyndromic, prelingual, profound hearing loss, we identified in the gene FAM65B (MIM611410) a splice site mutation (c.102-1G>A) that perfectly cosegregates with the phenotype in the family. The mutation leads to exon skipping and deletion of 52-amino acid residues of a PX membrane localization domain. FAM65B is known to be involved in myotube formation and in regulation of cell adhesion, polarization, and migration. We show that wild-type Fam65b is expressed during embryonic and postnatal development stages in murine cochlea, and that the protein localizes to the plasma membranes of the stereocilia of inner and outer hair cells of the inner ear. The wild-type protein targets the plasma membrane, whereas the mutant protein accumulates in cytoplasmic inclusion bodies and does not reach the membrane. In zebrafish, knockdown of fam65b leads to significant reduction of numbers of saccular hair cells and neuromasts and to hearing loss. We conclude that FAM65B is a plasma membrane-associated protein of hair cell stereocilia that is essential for hearing.


Asunto(s)
Audición/fisiología , Proteínas/fisiología , Estereocilios/fisiología , Animales , Moléculas de Adhesión Celular , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Audición/genética , Pérdida Auditiva Sensorineural/genética , Humanos , Masculino , Ratones , Linaje , Proteínas/genética , Proteínas/metabolismo , Empalme del ARN , Fracciones Subcelulares/metabolismo , Turquía , Pez Cebra
18.
Ann Hum Genet ; 80(6): 327-331, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27870113

RESUMEN

Identifying the genetic etiology in a person with hearing loss (HL) is challenging due to the extreme genetic heterogeneity in HL and the population-specific variability. In this study, after excluding GJB2 variants, targeted resequencing of 180 deafness-related genes revealed the causative variants in 11 of 19 (58%) Brazilian probands with autosomal recessive HL. Identified pathogenic variants were in MYO15A (10 families) and CLDN14 (one family). Remarkably, the MYO15A p.(Val1400Met) variant was identified in eight families from the city of Monte Santo in the northeast region of Brazil. Haplotype analysis of this variant was consistent with a single founder. No other cases with this variant were detected among 105 simplex cases from other cities of northeastern Brazil, suggesting that this variant is confined to a geographical region. This study suggests that it is feasible to develop population-specific screening for deafness variants once causative variants are identified in different geographical groups.


Asunto(s)
Pérdida Auditiva/genética , Miosinas/genética , Brasil , Estudios de Casos y Controles , Claudinas/genética , Análisis Mutacional de ADN , Efecto Fundador , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Mutación Missense
19.
Hum Genet ; 135(8): 953-61, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27344577

RESUMEN

Hearing loss is the most common sensory deficit in humans with causative variants in over 140 genes. With few exceptions, however, the population-specific distribution for many of the identified variants/genes is unclear. Until recently, the extensive genetic and clinical heterogeneity of deafness precluded comprehensive genetic analysis. Here, using a custom capture panel (MiamiOtoGenes), we undertook a targeted sequencing of 180 genes in a multi-ethnic cohort of 342 GJB2 mutation-negative deaf probands from South Africa, Nigeria, Tunisia, Turkey, Iran, India, Guatemala, and the United States (South Florida). We detected causative DNA variants in 25 % of multiplex and 7 % of simplex families. The detection rate varied between 0 and 57 % based on ethnicity, with Guatemala and Iran at the lower and higher end of the spectrum, respectively. We detected causative variants within 27 genes without predominant recurring pathogenic variants. The most commonly implicated genes include MYO15A, SLC26A4, USH2A, MYO7A, MYO6, and TRIOBP. Overall, our study highlights the importance of family history and generation of databases for multiple ethnically discrete populations to improve our ability to detect and accurately interpret genetic variants for pathogenicity.


Asunto(s)
Sordera/genética , Genética de Población , Síndromes de Usher/genética , Sordera/epidemiología , Etnicidad/genética , Femenino , Pruebas Genéticas , Humanos , Masculino , Mutación , Síndromes de Usher/epidemiología
20.
Genet Med ; 18(4): 364-71, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26226137

RESUMEN

PURPOSE: Autosomal recessive nonsyndromic deafness (ARNSD) is characterized by a high degree of genetic heterogeneity, with reported mutations in 58 different genes. This study was designed to detect deafness-causing variants in a multiethnic cohort with ARNSD by using whole-exome sequencing (WES). METHODS: After excluding mutations in the most common gene, GJB2, we performed WES in 160 multiplex families with ARNSD from Turkey, Iran, Mexico, Ecuador, and Puerto Rico to screen for mutations in all known ARNSD genes. RESULTS: We detected ARNSD-causing variants in 90 (56%) families, 54% of which had not been previously reported. Identified mutations were located in 31 known ARNSD genes. The most common genes with mutations were MYO15A (13%), MYO7A (11%), SLC26A4 (10%), TMPRSS3 (9%), TMC1 (8%), ILDR1 (6%), and CDH23 (4%). Nine mutations were detected in multiple families with shared haplotypes, suggesting founder effects. CONCLUSION: We report on a large multiethnic cohort with ARNSD in which comprehensive analysis of all known ARNSD genes identifies causative DNA variants in 56% of the families. In the remaining families, WES allows us to search for causative variants in novel genes, thus improving our ability to explain the underlying etiology in more families.Genet Med 18 4, 364-371.


Asunto(s)
Sordera/diagnóstico , Sordera/genética , Exoma , Genes Recesivos , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Alelos , Estudios de Cohortes , Etnicidad/genética , Genotipo , Humanos , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA