Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 21(14)2020 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-32650483

RESUMEN

While a plethora of genetic techniques have been developed over the past century, modifying specific sequences of the fruit fly genome has been a difficult, if not impossible task. clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 truly redefined molecular genetics and provided new tools to model human diseases in Drosophila melanogaster. This is particularly true for genes whose protein sequences are highly conserved. Phosphoribosyl pyrophosphate synthetase (PRPS) is a rate-limiting enzyme in nucleotide metabolism whose missense mutations are found in several neurological disorders, including Arts syndrome. In addition, PRPS is deregulated in cancer, particularly those that become resistant to cancer therapy. Notably, Drosophila PRPS shares about 90% protein sequence identity with its human orthologs, making it an ideal gene to study via CRISPR/Cas9. In this review, we will summarize recent findings on PRPS mutations in human diseases including cancer and on the molecular mechanisms by which PRPS activity is regulated. We will also discuss potential applications of Drosophila CRISPR/Cas9 to model PRPS-dependent disorders and other metabolic diseases that are associated with nucleotide metabolism.


Asunto(s)
Ataxia/genética , Trastornos Sordoceguera/genética , Drosophila melanogaster/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Ribosa-Fosfato Pirofosfoquinasa/genética , Animales , Sistemas CRISPR-Cas/genética , Modelos Animales de Enfermedad , Edición Génica/métodos , Humanos , Mutación/genética
2.
Hum Mol Genet ; 26(6): 1157-1172, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28137943

RESUMEN

The Usher syndrome (USH) is the most common form of inherited deaf-blindness, accompanied by vestibular dysfunction. Due to the heterogeneous manifestation of the clinical symptoms, three USH types (USH1-3) and additional atypical forms are distinguished. USH1 and USH2 proteins have been shown to function together in multiprotein networks in photoreceptor cells and hair cells. Mutations in USH proteins are considered to disrupt distinct USH protein networks and finally lead to the development of USH.To get novel insights into the molecular pathomechanisms underlying USH, we further characterize the periciliary USH protein network in photoreceptor cells. We show the direct interaction between the scaffold protein SANS (USH1G) and the transmembrane adhesion protein ush2a and that both assemble into a ternary USH1/USH2 complex together with the PDZ-domain protein whirlin (USH2D) via mutual interactions. Immunohistochemistry and proximity ligation assays demonstrate co-localization of complex partners and complex formation, respectively, in the periciliary region, the inner segment and at the synapses of rodent and human photoreceptor cells. Protein-protein interaction assays and co-expression of complex partners reveal that pathogenic mutations in USH1G severely affect formation of the SANS/ush2a/whirlin complex. Translational read-through drug treatment, targeting the c.728C > A (p.S243X) nonsense mutation, restored SANS scaffold function. We conclude that USH1 and USH2 proteins function together in higher order protein complexes. The maintenance of USH1/USH2 protein complexes depends on multiple USH1/USH2 protein interactions, which are disrupted by pathogenic mutations in USH1G protein SANS.


Asunto(s)
Trastornos Sordoceguera/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Síndromes de Usher/genética , Trastornos Sordoceguera/patología , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/metabolismo , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Humanos , Proteínas de la Membrana/química , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutación , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Unión Proteica , Mapas de Interacción de Proteínas/genética , Estructura Terciaria de Proteína , Síndromes de Usher/complicaciones , Síndromes de Usher/patología
3.
BMC Med Genet ; 20(1): 68, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31046701

RESUMEN

BACKGROUND: Usher syndrome, the most common form of inherited deaf-blindness, is unlike many other forms of syndromic hereditary hearing loss in that the extra aural clinical manifestations are also detrimental to communication. Usher syndrome patients with early onset deafness also experience vision loss due to progressive retinitis pigmentosa that can lead to legal blindness in their third or fourth decade. METHODS: Using a multi-omic approach, we identified three novel pathogenic variants in two Usher syndrome genes (USH2A and ADGRV1) in cases initially referred for isolated vision or hearing loss. RESULTS: In a multiplex hearing loss family, two affected sisters, the product of a second cousin union, are homozygous for a novel nonsense pathogenic variant in ADGRV1 (c.17062C > T, p.Arg5688*), predicted to create a premature stop codon near the N-terminus of ADGRV1. Ophthalmological examination of the sisters confirmed typical retinitis pigmentosa and prompted a corrected Usher syndrome diagnosis. In an unrelated clinical case, a child with hearing loss tested positive for two novel USH2A splicing variants (c.5777-1G > A, p. Glu1926_Ala1952del and c.10388-2A > G, p.Asp3463Alafs*6) and RNA studies confirmed that both pathogenic variants cause splicing errors. Interestingly, these same USH2A variants are also identified in another family with vision loss where subsequent clinical follow-up confirmed pre-existing hearing loss since early childhood, eventually resulting in a reassigned diagnosis of Usher syndrome. CONCLUSION: These findings provide empirical evidence to increase Usher syndrome surveillance of at-risk children. Given that novel antisense oligonucleotide therapies have been shown to rescue retinal degeneration caused by USH2A splicing pathogenic variants, these solved USH2A patients may now be eligible to be enrolled in therapeutic trials.


Asunto(s)
Trastornos Sordoceguera/genética , Síndromes de Usher/genética , Niño , Preescolar , Femenino , Genotipo , Humanos , Masculino , Linaje , Fenotipo
4.
BMC Med Genet ; 20(1): 11, 2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30634948

RESUMEN

BACKGROUND: Mohr-Tranebjaerg syndrome (MTS) is a rare X-linked recessive neurodegenerative disorder resulting in early-onset hearing impairment, gradual dystonia and optic atrophy. MTS is caused by variations in the nuclear TIMM8A gene, which is involved in mitochondrial transport of metabolites. This study aimed to identify the pathogenic gene variations in three Chinese families associated with predicted MTS with or without X-linked agammaglobulinaemia. METHODS: Otologic examinations, vestibular, neurological, optical and other clinical evaluations were conducted on the family members. Targeted genes capture combining next generation sequencing (NGS) was performed, and then Sanger sequencing was used to confirm the causative variation. RESULTS: A novel variation, c.232_233insCAAT, in TIMM8A was identified as the pathogenic variation in one Chinese family. This variation co-segregated with the most frequent phenotypic deafness and was absent in the 1000 Genomes Project, ExAC and 1751 ethnicity-matched controls. Clinically, otological examinations illustrated the typical postsynaptic auditory neuropathy for the proband without the symptoms of dystonia or optic atrophy. MRI demonstrated abnormal small cochlear symmetric nerves, while the vestibular function appeared to be less influenced. Furthermore, we found another two TIMM8A variations, the deletion c.133_135delGAG and a copy number variation (CNV) including the TIMM8A gene, in two independent case, when we performed NGS on an auditory neuropathy population. CONCLUSION: We identified two novel variations in the TIMM8A gene (c.232_233insCAAT and c.133_135delGAG) and a CNV including the TIMM8A gene in three independent Chinese families with predicted MTS. To our knowledge, this is the first report of TIMM8A variations being identified in a Chinese population. Our results enrich the variation spectrum of TIMM8A and clinical heterogeneity of MTS. Genetic detection and diagnosis is a powerful tool for better understanding and managing syndromic hearing impairments, such as MTS, before they become full-blown.


Asunto(s)
Trastornos Sordoceguera/diagnóstico , Trastornos Sordoceguera/genética , Distonía/diagnóstico , Distonía/genética , Pruebas Genéticas/métodos , Pérdida Auditiva Central/diagnóstico , Pérdida Auditiva Central/genética , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Proteínas de Transporte de Membrana/genética , Atrofia Óptica/diagnóstico , Atrofia Óptica/genética , Fenotipo , Agammaglobulinemia/genética , Pueblo Asiatico/genética , Variaciones en el Número de Copia de ADN , Sordera/genética , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Variación Genética , Humanos , Masculino , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Mutación , Linaje
5.
Ann Neurol ; 82(6): 1004-1015, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29205472

RESUMEN

OBJECTIVE: 3-Methylglutaconic aciduria, dystonia-deafness, hepatopathy, encephalopathy, Leigh-like syndrome (MEGDHEL) syndrome is caused by biallelic variants in SERAC1. METHODS: This multicenter study addressed the course of disease for each organ system. Metabolic, neuroradiological, and genetic findings are reported. RESULTS: Sixty-seven individuals (39 previously unreported) from 59 families were included (age range = 5 days-33.4 years, median age = 9 years). A total of 41 different SERAC1 variants were identified, including 20 that have not been reported before. With the exception of 2 families with a milder phenotype, all affected individuals showed a strikingly homogeneous phenotype and time course. Severe, reversible neonatal liver dysfunction and hypoglycemia were seen in >40% of all cases. Starting at a median age of 6 months, muscular hypotonia (91%) was seen, followed by progressive spasticity (82%, median onset = 15 months) and dystonia (82%, 18 months). The majority of affected individuals never learned to walk (68%). Seventy-nine percent suffered hearing loss, 58% never learned to speak, and nearly all had significant intellectual disability (88%). Magnetic resonance imaging features were accordingly homogenous, with bilateral basal ganglia involvement (98%); the characteristic "putaminal eye" was seen in 53%. The urinary marker 3-methylglutaconic aciduria was present in virtually all patients (98%). Supportive treatment focused on spasticity and drooling, and was effective in the individuals treated; hearing aids or cochlear implants did not improve communication skills. INTERPRETATION: MEGDHEL syndrome is a progressive deafness-dystonia syndrome with frequent and reversible neonatal liver involvement and a strikingly homogenous course of disease. Ann Neurol 2017;82:1004-1015.


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Trastornos Sordoceguera/diagnóstico por imagen , Trastornos Sordoceguera/genética , Progresión de la Enfermedad , Distonía/diagnóstico por imagen , Distonía/genética , Discapacidad Intelectual/diagnóstico por imagen , Discapacidad Intelectual/genética , Mutación/genética , Atrofia Óptica/diagnóstico por imagen , Atrofia Óptica/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Niño , Preescolar , Estudios de Cohortes , Trastornos Sordoceguera/terapia , Distonía/terapia , Femenino , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/terapia , Masculino , Atrofia Óptica/terapia , Adulto Joven
6.
Metab Brain Dis ; 33(2): 589-600, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29047041

RESUMEN

Charcot-Marie-Tooth disease (CMT) is one of the most commonly inherited congenital neurological disorders, affecting approximately 1 in 2500 in the US. About 80 genes were found to be in association with CMT. The phosphoribosyl pyrophosphate synthetase 1 (PRPS1) is an essential enzyme in the primary stage of de novo and salvage nucleotide synthesis. The mutations in the PRPS1 gene leads to X-linked Charcot-Marie-Tooth neuropathy type 5 (CMTX5), PRS super activity, Arts syndrome, X-linked deafness-1, breast cancer, and colorectal cancer. In the present study, we obtained 20 missense mutations from UniProt and dbSNP databases and applied series of comprehensive in silico prediction methods to assess the degree of pathogenicity and stability. In silico tools predicted four missense mutations (D52H, M115 T, L152P, and D203H) to be potential disease causing mutations. We further subjected the four mutations along with native protein to 50 ns molecular dynamics simulation (MDS) using Gromacs package. The resulting trajectory files were analyzed to understand the stability differences caused by the mutations. We used the Root Mean Square Deviation (RMSD), Radius of Gyration (Rg), solvent accessibility surface area (SASA), Covariance matrix, Principal Component Analysis (PCA), Free Energy Landscape (FEL), and secondary structure analysis to assess the structural changes in the protein upon mutation. Our study suggests that the four mutations might affect the PRPS1 protein function and stability of the structure. The proposed study may serve as a platform for drug repositioning and personalized medicine for diseases that are caused by the PRPS1 deficiency.


Asunto(s)
Ataxia/genética , Enfermedad de Charcot-Marie-Tooth/genética , Trastornos Sordoceguera/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Mutación/genética , Ribosa-Fosfato Pirofosfoquinasa/deficiencia , Secuencia de Aminoácidos , Enfermedad de Charcot-Marie-Tooth/diagnóstico , Humanos , Fenotipo , Ribosa-Fosfato Pirofosfoquinasa/genética
7.
Klin Monbl Augenheilkd ; 235(3): 273-280, 2018 Mar.
Artículo en Alemán | MEDLINE | ID: mdl-29534264

RESUMEN

The human Usher syndrome (USH) is a complex, rare disease manifesting in its most common form of inherited deaf-blindness. Due to the heterogeneous manifestation of the clinical symptoms, three clinical types (USH1-3) are distinguished according to the severity of the disease pattern. For a correct diagnosis, in addition to the auditory tests in early newborn screening, ophthalmological examinations and molecular genetic analysis are important. Ten known USH genes encode proteins, which are from heterogeneous protein families, interact in functional protein networks. In the eye and in the ear, USH proteins are expressed primarily in the mechano-sensitive hair cells and the rod and cone photoreceptor cells, respectively. In the hair cells, the USH protein networks are essential for the correct differentiation of the hair bundles as well as for the function of the mechano-electrical transduction complex in the matured cell. In the photoreceptor cells, USH proteins are located in the ciliary region and participate in intracellular transport processes. In addition, a USH protein network is present in the so-called calyceal processes. The lack of calyceal processes and the absence of a prominent visual phenotype in the mouse disqualifies mice as models for studies on the ophthalmic component of USH. While hearing impairments can be compensated with hearing aids and cochlear implants, there is no practical therapy for USH in the eye. Currently, gene-based therapy concepts, such as gene addition, applications of antisense oligonucleotides and TRIDs ("translational readthrough inducing drugs") for the readthrough of nonsense mutations are preclinically evaluated. For USH1B/MYO7A the UshStat gene therapy clinical trial is ongoing.


Asunto(s)
Ciliopatías/diagnóstico , Enfermedades Raras , Síndromes de Usher/diagnóstico , Animales , Ciliopatías/clasificación , Ciliopatías/genética , Ciliopatías/terapia , Análisis Mutacional de ADN , Trastornos Sordoceguera/clasificación , Trastornos Sordoceguera/diagnóstico , Trastornos Sordoceguera/genética , Trastornos Sordoceguera/terapia , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Ratones , Tamizaje Neonatal , Células Fotorreceptoras de Vertebrados/fisiología , Embarazo , Síndromes de Usher/clasificación , Síndromes de Usher/genética , Síndromes de Usher/terapia
8.
Mov Disord ; 32(1): 162-165, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27862284

RESUMEN

INTRODUCTION: Dystonia-deafness syndrome is a distinct clinical presentation within the dystonia-spectrum. Although several genetic and acquired causes have been reported, etiology remains unknown in the majority of patients. OBJECTIVES: To describe two patients with dystonia-deafness syndrome due to a beta-actin gene mutation. METHODS: We report on disease course, genetic testing, and management of 2 patients, mother and daughter, presenting with dystonia-deafness syndrome. RESULTS: After exclusion of known dystonia-deafness syndrome causes, whole-exome sequencing revealed a beta-actin gene mutation (p.Arg183Trp) in both patients. Although beta-actin gene mutations are generally associated with developmental Baraitser-Winter syndrome, dystonia-deafness syndrome has been reported once in identical twin brothers. Bilateral GPi-DBS led to a significant decrease of dystonia and regain of independency in our patients. CONCLUSION: The p.Arg183Trp mutation in the beta-actin gene is associated with the clinical presentation of dystonia-deafness syndrome, even with only minimal or no developmental abnormalities of Baraitser-Winter syndrome. GPi-DBS should be considered to ameliorate the invalidating dystonia in these patients. © 2016 International Parkinson and Movement Disorder Society.


Asunto(s)
Actinas/genética , Trastornos Sordoceguera/genética , Trastornos Sordoceguera/terapia , Estimulación Encefálica Profunda/métodos , Distonía/genética , Distonía/terapia , Discapacidad Intelectual/genética , Discapacidad Intelectual/terapia , Atrofia Óptica/genética , Atrofia Óptica/terapia , Adulto , Femenino , Humanos , Persona de Mediana Edad , Madres , Mutación , Núcleo Familiar , Adulto Joven
9.
Hum Mutat ; 37(2): 170-4, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26593283

RESUMEN

Deafblindness is part of several genetic disorders. We investigated a consanguineous Egyptian family with two siblings affected by congenital hearing loss and retinal degeneration, initially diagnosed as Usher syndrome type 1. At teenage, severe enamel dysplasia, developmental delay, and microcephaly became apparent. Genome-wide homozygosity mapping and whole-exome sequencing detected a homozygous missense mutation, c.1238G>T (p.Gly413Val), affecting a highly conserved residue of peroxisomal biogenesis factor 6, PEX6. Biochemical profiling of the siblings revealed abnormal and borderline plasma phytanic acid concentration, and cerebral imaging revealed white matter disease in both. We show that Pex6 localizes to the apical extensions of secretory ameloblasts and differentiated odontoblasts at early stages of dentin synthesis in mice, and to cilia of retinal photoreceptor cells. We propose PEX6, and possibly other peroxisomal genes, as candidate for the rare cooccurrence of deafblindness and enamel dysplasia. Our study for the first time links peroxisome biogenesis disorders to retinal ciliopathies.


Asunto(s)
Adenosina Trifosfatasas/genética , Trastornos Sordoceguera/genética , Hipoplasia del Esmalte Dental/genética , Microcefalia/genética , Mutación Missense , Degeneración Retiniana/genética , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/metabolismo , Ameloblastos/metabolismo , Ameloblastos/patología , Secuencia de Aminoácidos , Animales , Niño , Cilios/metabolismo , Cilios/patología , Consanguinidad , Trastornos Sordoceguera/metabolismo , Trastornos Sordoceguera/patología , Hipoplasia del Esmalte Dental/metabolismo , Hipoplasia del Esmalte Dental/patología , Femenino , Expresión Génica , Homocigoto , Humanos , Masculino , Ratones , Microcefalia/metabolismo , Microcefalia/patología , Datos de Secuencia Molecular , Odontoblastos/metabolismo , Odontoblastos/patología , Linaje , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Hermanos , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Adulto Joven
10.
PLoS Genet ; 8(4): e1002671, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22532808

RESUMEN

Many aspects of behavior and physiology are under circadian control. In Drosophila, the molecular clock that regulates rhythmic patterns of behavior has been extensively characterized. In contrast, genetic loci involved in linking the clock to alterations in motor activity have remained elusive. In a forward-genetic screen, we uncovered a new component of the circadian output pathway, which we have termed dyschronic (dysc). dysc mutants exhibit arrhythmic locomotor behavior, yet their eclosion rhythms are normal and clock protein cycling remains intact. Intriguingly, dysc is the closest Drosophila homolog of whirlin, a gene linked to type II Usher syndrome, the leading cause of deaf-blindness in humans. Whirlin and other Usher proteins are expressed in the mammalian central nervous system, yet their function in the CNS has not been investigated. We show that DYSC is expressed in major neuronal tracts and regulates expression of the calcium-activated potassium channel SLOWPOKE (SLO), an ion channel also required in the circadian output pathway. SLO and DYSC are co-localized in the brain and control each other's expression post-transcriptionally. Co-immunoprecipitation experiments demonstrate they form a complex, suggesting they regulate each other through protein-protein interaction. Furthermore, electrophysiological recordings of neurons in the adult brain show that SLO-dependent currents are greatly reduced in dysc mutants. Our work identifies a Drosophila homolog of a deaf-blindness gene as a new component of the circadian output pathway and an important regulator of ion channel expression, and suggests novel roles for Usher proteins in the mammalian nervous system.


Asunto(s)
Encéfalo , Ritmo Circadiano/genética , Drosophila melanogaster , Neuronas , Animales , Conducta Animal , Encéfalo/metabolismo , Trastornos Sordoceguera/genética , Trastornos Sordoceguera/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Proteínas de la Membrana , Actividad Motora/genética , Neuronas/metabolismo , Neuronas/fisiología , Mapas de Interacción de Proteínas/genética
11.
Ann Otol Rhinol Laryngol ; 124 Suppl 1: 77S-83S, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25743180

RESUMEN

OBJECTIVE: This study examines ABHD12 mutation analysis in 2 PHARC patients, originally thought to be Usher syndrome. METHODS: The ABHD12 gene of 2 patients, who suffered from deaf-blindness and dysfunctional central and peripheral nervous systems, were sequenced. RESULTS: We identified that both cases carried the same novel splice site mutation in the ABHD12 gene. However, 1 had epilepsy and the other had peripheral neuropathy. Based on haplotype analysis, the mutation is likely not a hot spot, but rather could be attributable to a common ancestor. CONCLUSION: This study shows that PHARC has phenotypic variability, even within a family, which is consistent with previous reports. Differential diagnosis of "deaf-blindness" diseases is crucial. Confirming the presence of associated symptoms is necessary for differentiating some deaf-blindness syndromes. In addition, mutation analysis is a useful tool for confirming the diagnosis.


Asunto(s)
Ataxia/genética , Catarata/genética , Trastornos Sordoceguera/genética , Monoacilglicerol Lipasas/genética , Polineuropatías/genética , Retinitis Pigmentosa/genética , Encéfalo/patología , Análisis Mutacional de ADN , Trastornos Sordoceguera/diagnóstico , Diagnóstico Diferencial , Humanos , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Síndromes de Usher/genética
12.
Biochem Biophys Res Commun ; 430(2): 585-91, 2013 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23219819

RESUMEN

Mitochondrial diseases are a clinically heterogeneous group of disorders that arise as a result of dysfunction of the mitochondrial respiratory chain. Sensorineural hearing loss (SNHL) has been described in association to different mitochondrial multisystem syndromes, often involving the central nervous system, neuromuscular, or endocrine organs. In this study, we described a Tunisian young girl with hearing impairment, congenital visual loss and maternally inherited diabetes. No mutation was found in the mitochondrial tRNA(Leu(UUR)) and the 12S rRNA genes. However, we detected the m.7444G>A mutation in the mitochondrial COI/tRNA(Ser(UCN)) genes. This mutation eliminates the termination codon of the MT-CO1 gene and extends the COI polypeptide by three amino acids (Lys-Gln-Lys) to the C-terminal. The whole mitochondrial genome screening revealed the presence of a novel mutation m.6498C>A (L199I) in the mitochondrial DNA-encoded subunit I of the cytochrome c oxidase (COX). This "probably damaging" transversion affects a highly conserved domain and it was absent in 200 Tunisian controls. The studied patient was classified under the haplogroup H2a.


Asunto(s)
Codón de Terminación/genética , Trastornos Sordoceguera/genética , Diabetes Mellitus/genética , Complejo IV de Transporte de Electrones/genética , Pérdida Auditiva Sensorineural/genética , Enfermedades Mitocondriales/genética , ARN de Transferencia de Serina/genética , Adolescente , Secuencia de Aminoácidos , Complejo IV de Transporte de Electrones/química , Femenino , Humanos , Datos de Secuencia Molecular , Mutación , Estructura Secundaria de Proteína , Túnez
14.
Mov Disord ; 28(6): 795-803, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23418071

RESUMEN

The syndrome of deafness-dystonia is rare and refers to the association of hearing impairment and dystonia when these are dominant features of a disease. Known genetic causes include Mohr-Tranebjaerg syndrome, Woodhouse-Sakati syndrome, and mitochondrial disorders, but the cause frequently remains unidentified. The aim of the current study was to better characterize etiological and clinical aspects of deafness-dystonia syndrome. We evaluated 20 patients with deafness-dystonia syndrome who were seen during the period between 1994 and 2011. The cause was identified in only 7 patients and included methylmalonic aciduria, meningoencephalitis, perinatal hypoxic-ischemic injury, large genomic deletion on chromosome 7q21, translocase of inner mitochondrial membrane 8 homolog A (TIMM8A) mutation (Mohr-Tranebjaerg syndrome), and chromosome 2 open reading frame 37 (C2orf37) mutation (Woodhouse-Sakati syndrome). The age of onset and clinical characteristics in these patients varied, depending on the etiology. In 13 patients, the cause remained unexplained despite extensive work-up. In the group of patients who had unknown etiology, a family history for deafness and/or dystonia was present the majority of patients, suggesting a strong genetic component. Sensory-neural deafness always preceded dystonia. Two clinical patterns of deafness-dystonia syndrome were observed: patients who had an onset in childhood had generalized dystonia (10 of 13 patients) with frequent bulbar involvement, whereas patients who had a dystonia onset in adulthood had segmental dystonia (3 of 13 patients) with the invariable presence of laryngeal dystonia. Deafness-dystonia syndrome is etiologically and clinically heterogeneous, and most patients have an unknown cause. The different age at onset and variable family history suggest a heterogeneous genetic background, possibly including currently unidentified genetic conditions.


Asunto(s)
Trastornos Sordoceguera/genética , Distonía/genética , Heterogeneidad Genética , Discapacidad Intelectual/genética , Proteínas de Transporte de Membrana/genética , Mutación/genética , Proteínas Nucleares/genética , Atrofia Óptica/genética , Adolescente , Adulto , Edad de Inicio , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al ADN/genética , Trastornos Sordoceguera/etiología , Progresión de la Enfermedad , Distonía/etiología , Salud de la Familia , Femenino , Pruebas Genéticas , Humanos , Discapacidad Intelectual/etiología , Leviviridae , Masculino , Persona de Mediana Edad , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Atrofia Óptica/etiología , Estudios Retrospectivos , Complejos de Ubiquitina-Proteína Ligasa , Adulto Joven
15.
Genes (Basel) ; 14(3)2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36980899

RESUMEN

Cochlear implantation (CI) for deafblindness may have more impact than for non-syndromic hearing loss. Deafblind patients have a double handicap in a society that is more and more empowered by fast communication. CI is a remedy for deafness, but requires revision surgery every 20 to 25 years, and thus placement should be minimally invasive. Furthermore, failed reimplantation surgery will have more impact on a deafblind person. In this context, we assessed the safety of minimally invasive robotically assisted cochlear implant surgery (RACIS) for the first time in a deafblind patient. Standard pure tone audiometry and speech audiometry were performed in a patient with deafblindness as part of this robotic-assisted CI study before and after surgery. This patient, with an optic atrophy 1 (OPA1) (OMIM#165500) mutation consented to RACIS for the second (contralateral) CI. The applicability and safety of RACIS were evaluated as well as her subjective opinion on her disability. RACIS was uneventful with successful surgical and auditory outcomes in this case of deafblindness due to the OPA1 mutation. RACIS appears to be a safe and beneficial intervention to increase communication skills in the cases of deafblindness due to an OPA1 mutation. The use of RACIS use should be widespread in deafblindness as it minimizes surgical trauma and possible failures.


Asunto(s)
Implantación Coclear , Implantes Cocleares , Trastornos Sordoceguera , Sordera , Femenino , Humanos , Implantación Coclear/métodos , Trastornos Sordoceguera/genética , Trastornos Sordoceguera/cirugía , Sordera/genética , Sordera/cirugía , GTP Fosfohidrolasas/genética , Mutación
16.
Mov Disord ; 27(8): 1034-40, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22736418

RESUMEN

Mohr-Tranebjaerg syndrome (MTS) is an X-linked recessive disorder characterized by deafness and dystonia. However the phenotypic expression of dystonia has not been systematically defined. We report clinical, neurophysiological, and ophthalmological data on 6 subjects from 3 Australian kindreds, including 2 with novel mutations, together with a systematic review of the literature, in order to define the phenotypic expression of dystonia. Profound hearing impairment in affected males develops by infancy and precedes the development of dystonia, which varies in time of onset from the first to the sixth decades, with a peak in the second and third decades. Dystonia in MTS tends to be focal, segmental, or multifocal in distribution at onset, with a predilection for the upper body, variably involving the head, neck, and upper limbs. The majority of patients have progression or generalization of their dystonia regardless of age of onset. Within our 3 kindreds, we observed relative intrafamilial homogeneity but interfamilial variation. The median time to the development of moderate-severely disabling dystonia in these subjects was 11 years. Associated features included progressive cognitive decline, pyramidal signs, and in 1 patient, gait freezing and postural instability. Optic atrophy and cortical visual impairment were both observed. We report for the first time a female patient who developed multiple disabling neurological complications of MTS. Our findings more clearly define and expand the phenotype of both the dystonia and other neurological features of MTS and have implications for the diagnosis and management of this condition.


Asunto(s)
Trastornos Sordoceguera/genética , Trastornos Sordoceguera/fisiopatología , Distonía/genética , Distonía/fisiopatología , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Atrofia Óptica/genética , Atrofia Óptica/fisiopatología , Adolescente , Adulto , Edad de Inicio , Australia , Niño , Preescolar , Trastornos del Conocimiento/etiología , Trastornos Sordoceguera/psicología , Progresión de la Enfermedad , Distonía/psicología , Potenciales Evocados Somatosensoriales , Potenciales Evocados Visuales , Exones , Femenino , Humanos , Lactante , Discapacidad Intelectual/etiología , Discapacidad Intelectual/psicología , Intrones , Imagen por Resonancia Magnética , Masculino , Proteínas de Transporte de Membrana/genética , Persona de Mediana Edad , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Mutación , Pruebas Neuropsicológicas , Atrofia Óptica/psicología , Linaje , Caracteres Sexuales , Adulto Joven
17.
Am J Med Genet A ; 158A(2): 455-60, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22246954

RESUMEN

We identified a novel missense mutation, c.424G>C (p.Val142Leu) in PRPS1 in a patient with uric acid overproduction without gout but with developmental delay, hypotonia, hearing loss, and recurrent respiratory infections. The uric acid overproduction accompanying this combination of symptoms suggests that the patient presented with phosphoribosylpyrophosphate (PRPP) synthetase superactivity, but recurrent infections have not been associated with superactivity until now. However, recurrent infections are a prominent feature of patients with Arts syndrome, which is caused by PRPS1 loss-of-function mutations, indicating that the patient reported here has an intermediate phenotype. Molecular modeling predicts that the p.Val142Leu change affects both allosteric sites that are involved in inhibition of PRPS1 and the ATP-binding site, which suggests that this substitution can result both in a gain-of-function and loss-of-function of PRPP synthetase. This finding is in line with the normal PRPP synthetase activity in fibroblasts and the absence of activity in erythrocytes of the present patient. We postulate that the overall effect of the p.Val142Leu change on protein activity is determined by the cell type, being a gain-of-function in proliferating cells and a loss-of-function in postmitotic cells. Our results show that missense mutations in PRPS1 can cause a continuous spectrum of features ranging from progressive non-syndromic postlingual hearing impairment to uric acid overproduction, neuropathy, and recurrent infections depending on the functional sites that are affected.


Asunto(s)
Ataxia/patología , Trastornos Sordoceguera/patología , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Infecciones/enzimología , Mutación Missense , Ribosa-Fosfato Pirofosfoquinasa/genética , Ribosa-Fosfato Pirofosfoquinasa/metabolismo , Ataxia/complicaciones , Ataxia/enzimología , Ataxia/genética , Preescolar , Trastornos Sordoceguera/complicaciones , Trastornos Sordoceguera/enzimología , Trastornos Sordoceguera/genética , Activación Enzimática/genética , Enfermedades Genéticas Ligadas al Cromosoma X/complicaciones , Enfermedades Genéticas Ligadas al Cromosoma X/enzimología , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Predisposición Genética a la Enfermedad , Pérdida Auditiva Bilateral/diagnóstico , Pérdida Auditiva Bilateral/patología , Humanos , Infecciones/complicaciones , Infecciones/patología , Modelos Moleculares , Hipotonía Muscular/diagnóstico , Hipotonía Muscular/patología , Mutación Missense/genética , Relación Estructura-Actividad , Ácido Úrico/sangre
18.
Sci Rep ; 12(1): 14959, 2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36056138

RESUMEN

Mohr-Tranebjærg syndrome is an X-linked syndrome characterized by sensorineural hearing impairment in childhood, followed by progressive neurodegeneration leading to a broad phenotypic spectrum. Genetically MTS is caused by pathogenic variants in the TIMM8A gene, including gene deletions and larger contiguous gene deletions. Some of the latter involve the neighboring gene BTK, resulting in agammaglobulinemia. By next-generation mate-pair sequencing we have mapped the chromosomal deletion breakpoints of one MTS case and three XLA-MTS cases and used breakpoint-spanning PCR to fine map the breakpoints by Sanger sequencing. Two of the XLA-MTS cases presented with large deletions (63.5 and 27.2 kb), and the junctional regions were characterized by long stretches of microhomology, indicating that the events have emerged through homologous recombination. Conversely, the MTS case exhibited a small 2 bp region of microhomology, and the regions were not characterized by extensive microhomology. The third XLA-MTS case had a more complex breakpoint, including a 59 bp inverted insertion, thus at least four breakpoints were involved in this event. In conclusion, mate-pair library generation combined with next-generation sequencing is an efficient method for breakpoint identification, also in regions characterized by repetitive elements.


Asunto(s)
Trastornos Sordoceguera , Distonía , Discapacidad Intelectual , Atrofia Óptica , Trastornos Sordoceguera/genética , Distonía/genética , Humanos , Discapacidad Intelectual/genética , Proteínas de Transporte de Membrana/genética , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Atrofia Óptica/genética
20.
J Hum Genet ; 56(8): 577-82, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21753765

RESUMEN

Patients with X-linked agammaglobulinemia (XLA) can present with sensorineural deafness. This can result from a gross deletion that not only involved the Bruton's tyrosine kinase (BTK) gene, but also TIMM8A, mutations in which underlie the Mohr-Tranebjærg syndrome (MTS). We analyzed the genomic break points observed in three XLA-MTS patients and compared these with deletions break points from XLA patients. Patient 1 had a 63-kb deletion with break points in intron 15 of BTK and 4 kb upstream of TAF7L. Patients 2 and 3 had 149.7 and 196 kb deletions comprising BTK, TIMM8A, TAF7L and DRP2. The break points in patients 1 and 3 were located in Alu and endogenous retrovirus (ERV) repeats, whereas the break points in patient 2 did not show involvement of transposable elements. Comparison of gross deletion sizes and involvement of transposable elements in XLA and XLA-MTS patients from the literature showed preferential involvement of Alu elements in smaller deletions (<10 kb). These results show further insights into the molecular mechanisms underlying gross deletions in patients with primary immunodeficiency.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos X/genética , Proteínas de Transporte de Membrana/genética , Proteínas Tirosina Quinasas/genética , Adolescente , Agammaglobulinemia Tirosina Quinasa , Agammaglobulinemia/genética , Elementos Alu/genética , Niño , Puntos de Rotura del Cromosoma , Trastornos Sordoceguera/genética , Distonía/genética , Eliminación de Gen , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Humanos , Lactante , Discapacidad Intelectual/genética , Masculino , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Atrofia Óptica/genética , Síndrome , Secuencias Repetidas Terminales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA