Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
EMBO Mol Med ; 15(10): e17393, 2023 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-37642150

RESUMEN

Deafness affects 5% of the world's population, yet there is a lack of treatments to prevent hearing loss due to genetic causes. Norrie disease is a recessive X-linked disorder, caused by NDP gene mutation. It manifests as blindness at birth and progressive sensorineural hearing loss, leading to debilitating dual sensory deprivation. To develop a gene therapy, we used a Norrie disease mouse model (Ndptm1Wbrg ), which recapitulates abnormal retinal vascularisation and progressive hearing loss. We delivered human NDP cDNA by intravenous injection of adeno-associated viral vector (AAV)9 at neonatal, juvenile and young adult pathological stages and investigated its therapeutic effects on the retina and cochlea. Neonatal treatment prevented the death of the sensory cochlear hair cells and rescued cochlear disease biomarkers as demonstrated by RNAseq and physiological measurements of auditory function. Retinal vascularisation and electroretinograms were restored to normal by neonatal treatment. Delivery of NDP gene therapy after the onset of the degenerative inner ear disease also ameliorated the cochlear pathology, supporting the feasibility of a clinical treatment for progressive hearing loss in people with Norrie disease.

2.
Arch Dis Child Fetal Neonatal Ed ; 108(5): 464-470, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36564163

RESUMEN

OBJECTIVE: To investigate individual and concomitant risk factors for hearing loss during neonatal care. DESIGN: Case-control study. SETTING: Community. POPULATION: 237 children born <32 weeks of gestation; 57 with hearing loss and 180 with normal hearing born between 2009 and 2013, matched for sex, gestation and year of birth. MAIN OUTCOME MEASURES: Data were abstracted from clinical records for overall risk factors daily for the first 14 days and then weekly until discharge from neonatal care. All infants were screened for the presence of m.1555A>G mutation. RESULTS: Children with hearing loss had lower birth weight for gestational age, more severe neonatal illness, with increased exposure to inotrope, steroid, gentamicin, vancomycin and furosemide, and more frequent physiological risk, elevated bilirubin and creatinine levels and acidosis, but no index child was found to have the m.1555A>G mutation, compared with one among controls. The duration of gentamicin, vancomycin or furosemide administration in the first 14 days was associated with impaired hearing (OR per dose: 1.25; 95% CI 1.14 to 1.38). Multivariate analyses revealed independent risks for hearing loss for each day when there was physiological risk (OR per day 1.15 (1.05 to 1.27)) and each day of medication exposure (OR 1.23 (1.1 to 1.37)). CONCLUSION: Although the relative contribution of underlying illness and medication cannot be identified by this study, cumulative use of ototoxic medication and the presence of physiologic risk factors independently increased the likelihood of hearing loss, warranting close monitoring of coincident therapy throughout neonatal critical care.


Asunto(s)
Sordera , Pérdida Auditiva , Lactante , Niño , Recién Nacido , Humanos , Recien Nacido Prematuro , Estudios de Casos y Controles , Vancomicina/efectos adversos , Furosemida/efectos adversos , Pérdida Auditiva/epidemiología , Pérdida Auditiva/inducido químicamente , Gentamicinas/efectos adversos , Factores de Riesgo
3.
Stem Cell Reports ; 17(11): 2421-2437, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36240775

RESUMEN

Usher syndrome-associated retinitis pigmentosa (RP) causes progressive retinal degeneration, which has no cure. The pathomechanism of Usher type 1B (USH1B)-RP caused by MYO7A mutation remains elusive because of the lack of faithful animal models and limited knowledge of MYO7A function. Here, we analyzed 3D retinal organoids generated from USH1B patient-derived induced pluripotent stem cells. Increased differential gene expression occurred over time without excessive photoreceptor cell death in USH1B organoids compared with controls. Dysregulated genes were enriched first for mitochondrial functions and then proteasomal ubiquitin-dependent protein catabolic processes and RNA splicing. Single-cell RNA sequencing revealed MYO7A expression in rod photoreceptor and Müller glial cells corresponding to upregulation of stress responses in NRL+ rods and apoptotic signaling pathways in VIM+ Müller cells, pointing to the defensive mechanisms that mitigate photoreceptor cell death. This first human model for USH1B-RP provides a representation of patient retina in vivo relevant for development of therapeutic strategies.


Asunto(s)
Organoides , Retinitis Pigmentosa , Animales , Humanos , Miosina VIIa , Organoides/patología , Patología Molecular , Miosinas/genética , Miosinas/metabolismo , Retina/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Células Fotorreceptoras Retinianas Bastones/patología
4.
Am J Ophthalmol ; 241: 9-27, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35469785

RESUMEN

OBJECTIVE: To evaluate the pattern of vision loss and genotype-phenotype correlations in WFS1-associated optic neuropathy (WON). DESIGN: Multicenter cohort study. METHODS: The study involved 37 patients with WON carrying pathogenic or candidate pathogenic WFS1 variants. Genetic and clinical data were retrieved from the medical records. Thirteen patients underwent additional comprehensive ophthalmologic assessment. Deep phenotyping involved visual electrophysiology and advanced psychophysical testing with a complementary metabolomic study. MAIN OUTCOME MEASURES: WFS1 variants, functional and structural optic nerve and retinal parameters, and metabolomic profile. RESULTS: Twenty-two recessive and 5 dominant WFS1 variants were identified. Four variants were novel. All WFS1 variants caused loss of macular retinal ganglion cells (RGCs) as assessed by optical coherence tomography (OCT) and visual electrophysiology. Advanced psychophysical testing indicated involvement of the major RGC subpopulations. Modeling of vision loss showed an accelerated rate of deterioration with increasing age. Dominant WFS1 variants were associated with abnormal reflectivity of the outer plexiform layer (OPL) on OCT imaging. The dominant variants tended to cause less severe vision loss compared with recessive WFS1 variants, which resulted in more variable phenotypes ranging from isolated WON to severe multisystem disease depending on the WFS1 alleles. The metabolomic profile included markers seen in other neurodegenerative diseases and type 1 diabetes mellitus. CONCLUSIONS: WFS1 variants result in heterogenous phenotypes influenced by the mode of inheritance and the disease-causing alleles. Biallelic WFS1 variants cause more variable, but generally more severe, vision and RGC loss compared with heterozygous variants. Abnormal cleftlike lamination of the OPL is a distinctive OCT feature that strongly points toward dominant WON.


Asunto(s)
Proteínas de la Membrana/genética , Enfermedades del Nervio Óptico , Estudios de Cohortes , Progresión de la Enfermedad , Estudios de Asociación Genética , Humanos , Nervio Óptico , Enfermedades del Nervio Óptico/diagnóstico , Enfermedades del Nervio Óptico/genética , Tomografía de Coherencia Óptica/métodos
5.
JCI Insight ; 7(3)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35132964

RESUMEN

Norrie disease is caused by mutation of the NDP gene, presenting as congenital blindness followed by later onset of hearing loss. Protecting patients from hearing loss is critical for maintaining their quality of life. This study aimed to understand the onset of pathology in cochlear structure and function. By investigating patients and juvenile Ndp-mutant mice, we elucidated the sequence of onset of physiological changes (in auditory brainstem responses, distortion product otoacoustic emissions, endocochlear potential, blood-labyrinth barrier integrity) and determined the cellular, histological, and ultrastructural events leading to hearing loss. We found that cochlear vascular pathology occurs earlier than previously reported and precedes sensorineural hearing loss. The work defines a disease mechanism whereby early malformation of the cochlear microvasculature precedes loss of vessel integrity and decline of endocochlear potential, leading to hearing loss and hair cell death while sparing spiral ganglion cells. This provides essential information on events defining the optimal therapeutic window and indicates that early intervention is needed. In an era of advancing gene therapy and small-molecule technologies, this study establishes Ndp-mutant mice as a platform to test such interventions and has important implications for understanding the progression of hearing loss in Norrie disease.


Asunto(s)
Ceguera/congénito , Manejo de la Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Predicción , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Pérdida Auditiva Sensorineural/fisiopatología , Audición/fisiología , Enfermedades del Sistema Nervioso/fisiopatología , Degeneración Retiniana/fisiopatología , Espasmos Infantiles/fisiopatología , Adolescente , Adulto , Animales , Ceguera/complicaciones , Ceguera/fisiopatología , Ceguera/terapia , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Estudios de Seguimiento , Enfermedades Genéticas Ligadas al Cromosoma X/complicaciones , Enfermedades Genéticas Ligadas al Cromosoma X/terapia , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/etiología , Humanos , Masculino , Ratones , Ratones Mutantes , Enfermedades del Sistema Nervioso/complicaciones , Enfermedades del Sistema Nervioso/terapia , Degeneración Retiniana/complicaciones , Degeneración Retiniana/terapia , Espasmos Infantiles/complicaciones , Espasmos Infantiles/terapia , Adulto Joven
6.
Sci Rep ; 10(1): 13763, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32792680

RESUMEN

Mutations in the SNX14 gene cause spinocerebellar ataxia, autosomal recessive 20 (SCAR20) in both humans and dogs. Studies implicating the phenotypic consequences of SNX14 mutations to be consequences of subcellular disruption to autophagy and lipid metabolism have been limited to in vitro investigation of patient-derived dermal fibroblasts, laboratory engineered cell lines and developmental analysis of zebrafish morphants. SNX14 homologues Snz (Drosophila) and Mdm1 (yeast) have also been conducted, demonstrated an important biochemical role during lipid biogenesis. In this study we report the effect of loss of SNX14 in mice, which resulted in embryonic lethality around mid-gestation due to placental pathology that involves severe disruption to syncytiotrophoblast cell differentiation. In contrast to other vertebrates, zebrafish carrying a homozygous, maternal zygotic snx14 genetic loss-of-function mutation were both viable and anatomically normal. Whilst no obvious behavioural effects were observed, elevated levels of neutral lipids and phospholipids resemble previously reported effects on lipid homeostasis in other species. The biochemical role of SNX14 therefore appears largely conserved through evolution while the consequences of loss of function varies between species. Mouse and zebrafish models therefore provide valuable insights into the functional importance of SNX14 with distinct opportunities for investigating its cellular and metabolic function in vivo.


Asunto(s)
Viabilidad Fetal/genética , Metabolismo de los Lípidos/genética , Placenta/anomalías , Nexinas de Clasificación/genética , Ataxias Espinocerebelosas/genética , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Desarrollo Embrionario/genética , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Fenotipo , Fosfolípidos/sangre , Embarazo , Trofoblastos/citología , Pez Cebra
7.
Hum Mol Genet ; 29(11): 1882-1899, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-31998945

RESUMEN

USH2A variants are the most common cause of Usher syndrome type 2, characterized by congenital sensorineural hearing loss and retinitis pigmentosa (RP), and also contribute to autosomal recessive non-syndromic RP. Several treatment strategies are under development; however, sensitive clinical trial endpoint metrics to determine therapeutic efficacy have not been identified. In the present study, we have performed longitudinal retrospective examination of the retinal and auditory symptoms in (i) 56 biallelic molecularly confirmed USH2A patients and (ii) ush2a mutant zebrafish to identify metrics for the evaluation of future clinical trials and rapid preclinical screening studies. The patient cohort showed a statistically significant correlation between age and both rate of constriction for the ellipsoid zone length and hyperautofluorescent outer retinal ring area. Visual acuity and pure tone audiograms are not suitable outcome measures. Retinal examination of the novel ush2au507 zebrafish mutant revealed a slowly progressive degeneration of predominantly rods, accompanied by rhodopsin and blue cone opsin mislocalization from 6 to 12 months of age with lysosome-like structures observed in the photoreceptors. This was further evaluated in the ush2armc zebrafish model, which revealed similar changes in photopigment mislocalization with elevated autophagy levels at 6 days post fertilization, indicating a more severe genotype-phenotype correlation and providing evidence of new insights into the pathophysiology underlying USH2A-retinal disease.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Pérdida Auditiva Sensorineural/genética , Retina/fisiopatología , Retinitis Pigmentosa/genética , Síndromes de Usher/genética , Adolescente , Adulto , Anciano , Animales , Autofagia/genética , Modelos Animales de Enfermedad , Electrorretinografía , Femenino , Estudios de Asociación Genética , Genotipo , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Opsinas/genética , Retina/diagnóstico por imagen , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Retinitis Pigmentosa/fisiopatología , Rodopsina/genética , Opsinas de Bastones/genética , Síndromes de Usher/diagnóstico por imagen , Síndromes de Usher/patología , Agudeza Visual/genética , Agudeza Visual/fisiología , Adulto Joven , Pez Cebra/genética
8.
Hum Mol Genet ; 28(22): 3815-3824, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31600780

RESUMEN

Wolfram syndrome (WS) is a heterogeneous multisystem neurodegenerative disorder with two allelic variations in addition to a separate subtype known as WS type 2. The wide phenotypic spectrum of WS includes diabetes mellitus and optic atrophy which is often accompanied by diabetes insipidus, deafness, urological and neurological complications in combination or in isolation. To date, the understanding of the genotype-phenotype relationship in this complex syndrome remains poorly understood. In this study, we identified and explored the functionality of rare and novel variants in the two causative WS genes WFS1 and CISD2 by assessing the effects of the mutations on the encoded proteins Wolframin and ERIS, in a cohort of 12 patients with autosomal recessive WS, dominant WS and WS type 2. The identified pathogenic variants included missense changes, frameshift deletions and insertions in WFS1 and an exonic deletion in CISD2 which all altered the respective encoded protein in a manner that did not correlate to the phenome previously described. These observations suggest the lack of genotype-phenotype correlation in this complex syndrome and the need to explore other molecular genetic mechanisms. Additionally, our findings highlight the importance of functionally assessing variants for their pathogenicity to tackle the problem of increasing variants of unknown significance in the public genetic databases.


Asunto(s)
Proteínas de la Membrana/genética , Síndrome de Wolfram/genética , Adolescente , Adulto , Alelos , Exones , Femenino , Mutación del Sistema de Lectura , Estudios de Asociación Genética , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Mutación , Atrofia Óptica/genética , Linaje , Fenotipo , Síndrome de Wolfram/fisiopatología
9.
Hum Mol Genet ; 28(20): 3466-3474, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31504499

RESUMEN

Recurrent pregnancy loss (RPL) is defined as two or more consecutive miscarriages and affects an estimated 1.5% of couples trying to conceive. RPL has been attributed to genetic, endocrine, immune and thrombophilic disorders, but many cases remain unexplained. We investigated a Bangladeshi family where the proband experienced 29 consecutive pregnancy losses with no successful pregnancies from three different marriages. Whole exome sequencing identified rare genetic variants in several candidate genes. These were further investigated in Asian and white European RPL cohorts, and in Bangladeshi controls. FKBP4, encoding the immunophilin FK506-binding protein 4, was identified as a plausible candidate, with three further novel variants identified in Asian patients. None were found in European patients or controls. In silico structural studies predicted damaging effects of the variants in the structure-function properties of the FKBP52 protein. These were located within domains reported to be involved in Hsp90 binding and peptidyl-prolyl cis-trans isomerase (PPIase) activity. Profound effects on PPIase activity were demonstrated in transiently transfected HEK293 cells comparing wild-type and mutant FKBP4 constructs. Mice lacking FKBP4 have been previously reported as infertile through implantation failure. This study therefore strongly implicates FKBP4 as associated with fetal losses in humans, particularly in the Asian population.


Asunto(s)
Aborto Habitual/genética , Secuenciación del Exoma/métodos , Proteínas de Unión a Tacrolimus/genética , Exoma/genética , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Mutación Missense/genética , Linaje , Embarazo , Estructura Secundaria de Proteína
10.
Genes (Basel) ; 10(7)2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31336982

RESUMEN

Labyrinthine aplasia, microtia, and microdontia (LAMM) is an autosomal recessive condition causing profound congenital deafness, complete absence of inner ear structures (usually Michel's aplasia), microtia (usually type 1) and microdontia. To date, several families have been described with this condition and a number of mutations has been reported. We report on eight further cases of LAMM syndrome including three novel mutations, c. 173T>C p.L58P; c. 284G>A p.(Arg95Gln) and c.325_327delinsA p.(Glu109Thrfs*18). Congenital deafness was the primary presenting feature in all affected individuals and consanguinity in all but two families. We compare the features in our patients to those previously reported in LAMM, and describe a milder, asymmetrical phenotype associated with FGF3 mutations.


Asunto(s)
Microtia Congénita/genética , Microtia Congénita/patología , Oído Interno/anomalías , Factor 3 de Crecimiento de Fibroblastos/genética , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Anomalías Dentarias/genética , Anomalías Dentarias/patología , Adulto , Preescolar , Consanguinidad , Análisis Mutacional de ADN , Sordera/congénito , Oído Interno/patología , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Fenotipo
11.
Science ; 364(6442)2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-31123110

RESUMEN

Approximately 2.4% of the human mitochondrial DNA (mtDNA) genome exhibits common homoplasmic genetic variation. We analyzed 12,975 whole-genome sequences to show that 45.1% of individuals from 1526 mother-offspring pairs harbor a mixed population of mtDNA (heteroplasmy), but the propensity for maternal transmission differs across the mitochondrial genome. Over one generation, we observed selection both for and against variants in specific genomic regions; known variants were more likely to be transmitted than previously unknown variants. However, new heteroplasmies were more likely to match the nuclear genetic ancestry as opposed to the ancestry of the mitochondrial genome on which the mutations occurred, validating our findings in 40,325 individuals. Thus, human mtDNA at the population level is shaped by selective forces within the female germ line under nuclear genetic control, which ensures consistency between the two independent genetic lineages.


Asunto(s)
ADN Mitocondrial/genética , Genoma Mitocondrial , Herencia Materna , Óvulo/crecimiento & desarrollo , Selección Genética , Femenino , Variación Genética , Humanos
12.
Ophthalmology ; 126(6): 888-907, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30653986

RESUMEN

PURPOSE: To develop a comprehensive next-generation sequencing panel assay that screens genes known to cause developmental eye disorders and inherited eye disease and to evaluate its diagnostic yield in a pediatric cohort with malformations of the globe, anterior segment anomalies, childhood glaucoma, or a combination thereof. DESIGN: Evaluation of diagnostic test. PARTICIPANTS: Two hundred seventy-seven children, 0 to 16 years of age, diagnosed with nonsyndromic or syndromic developmental eye defects without a genetic diagnosis. METHODS: We developed a new oculome panel using a custom-designed Agilent SureSelect QXT target capture method (Agilent Technologies, Santa Clara, CA) to capture and perform parallel high-throughput sequencing analysis of 429 genes associated with eye disorders. Bidirectional Sanger sequencing confirmed suspected pathogenic variants. MAIN OUTCOME MEASURES: Collated clinical details and oculome molecular genetic results. RESULTS: The oculome design covers 429 known eye disease genes; these are subdivided into 5 overlapping virtual subpanels for anterior segment developmental anomalies including glaucoma (ASDA; 59 genes), microphthalmia-anophthalmia-coloboma (MAC; 86 genes), congenital cataracts and lens-associated conditions (70 genes), retinal dystrophies (RET; 235 genes), and albinism (15 genes), as well as additional genes implicated in optic atrophy and complex strabismus (10 genes). Panel development and testing included analyzing 277 clinical samples and 3 positive control samples using Illumina sequencing platforms; more than 30× read depth was achieved for 99.5% of the targeted 1.77-Mb region. Bioinformatics analysis performed using a pipeline based on Freebayes and ExomeDepth to identify coding sequence and copy number variants, respectively, resulted in a definitive diagnosis in 68 of 277 samples, with variability in diagnostic yield between phenotypic subgroups: MAC, 8.2% (8 of 98 cases solved); ASDA, 24.8% (28 of 113 cases solved); other or syndromic, 37.5% (3 of 8 cases solved); RET, 42.8% (21 of 49 cases solved); and congenital cataracts and lens-associated conditions, 88.9% (8 of 9 cases solved). CONCLUSIONS: The oculome test diagnoses a comprehensive range of genetic conditions affecting the development of the eye, potentially replacing protracted and costly multidisciplinary assessments and allowing for faster targeted management. The oculome enabled molecular diagnosis of a significant number of cases in our sample cohort of varied ocular birth defects.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Anomalías del Ojo/diagnóstico , Anomalías del Ojo/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Técnicas de Diagnóstico Molecular , Mutación/genética , Proteoma/genética , Adolescente , Niño , Preescolar , Femenino , Genoma Humano , Humanos , Lactante , Recién Nacido , Masculino , Linaje
13.
Genet Med ; 21(5): 1083-1091, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30270361

RESUMEN

PURPOSE: Little is known about how health-care professionals communicate with patients about consenting to genome sequencing. We therefore examined what topics health-care professionals covered and what questions patients asked during consent conversations. METHODS: Twenty-one genome sequencing consent appointments were audio recorded and analyzed. Participants were 35 individuals being invited to participate in the 100,000 Genomes Project (14 participants with rare diseases, 21 relatives), and 10 health-care professionals ("consenters"). RESULTS: Two-thirds of participants' questions were substantive (e.g., genetics and inheritance); one-third administrative (e.g., filling in the consent form). Consenters usually (19/21) emphasized participant choice about secondary findings, but less often (13/21) emphasized the uncertainty about associated disease risks. Consenters primarily used passive statements and closed-ended, rather than open-ended, questions to invite participants' questions and concerns. In two appointments, one parent expressed negative or uncertain views about secondary findings, but after discussion with the other parent opted to receive them. CONCLUSION: Health-care professionals need to be prepared to answer patients' questions about genetics to facilitate genome sequencing consent. Health-care professionals' education also needs to address how to effectively listen and elicit each patient's questions and views, and how to discuss uncertainty around the disease risks associated with secondary findings.


Asunto(s)
Consentimiento Informado/ética , Secuenciación Completa del Genoma/ética , Adulto , Anciano , Comunicación , Formularios de Consentimiento/ética , Femenino , Conocimientos, Actitudes y Práctica en Salud , Personal de Salud , Humanos , Consentimiento Informado/normas , Masculino , Persona de Mediana Edad , Padres , Pacientes , Secuenciación Completa del Genoma/métodos
14.
J Med Genet ; 55(11): 721-728, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30049826

RESUMEN

BACKGROUND: Rare genetic conditions are frequent risk factors for, or direct causes of, paediatric intensive care unit (PICU) admission. Such conditions are frequently suspected but unidentified at PICU admission. Compassionate and effective care is greatly assisted by definitive diagnostic information. There is therefore a need to provide a rapid genetic diagnosis to inform clinical management.To date, whole genome sequencing (WGS) approaches have proved successful in diagnosing a proportion of children with rare diseases, but results may take months to report. Our aim was to develop an end-to-end workflow for the use of rapid WGS for diagnosis in critically ill children in a UK National Health Service (NHS) diagnostic setting. METHODS: We sought to establish a multidisciplinary Rapid Paediatric Sequencing team for case selection, trio WGS, rapid bioinformatics sequence analysis and a phased analysis and reporting system to prioritise genes with a high likelihood of being causal. RESULTS: Trio WGS in 24 critically ill children led to a molecular diagnosis in 10 (42%) through the identification of causative genetic variants. In 3 of these 10 individuals (30%), the diagnostic result had an immediate impact on the individual's clinical management. For the last 14 trios, the shortest time taken to reach a provisional diagnosis was 4 days (median 8.5 days). CONCLUSION: Rapid WGS can be used to diagnose and inform management of critically ill children within the constraints of an NHS clinical diagnostic setting. We provide a robust workflow that will inform and facilitate the rollout of rapid genome sequencing in the NHS and other healthcare systems globally.


Asunto(s)
Enfermedad Crítica , Enfermedades Genéticas Congénitas/diagnóstico , Enfermedades Genéticas Congénitas/genética , Secuenciación Completa del Genoma , Niño , Manejo de la Enfermedad , Estudio de Asociación del Genoma Completo/métodos , Estudio de Asociación del Genoma Completo/normas , Humanos , Unidades de Cuidado Intensivo Pediátrico , Enfermedades Raras , Secuenciación Completa del Genoma/métodos , Flujo de Trabajo
15.
Am J Hum Genet ; 102(6): 1195-1203, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29861108

RESUMEN

Next-generation sequencing is a powerful tool for the discovery of genes related to neurodevelopmental disorders (NDDs). Here, we report the identification of a distinct syndrome due to de novo or inherited heterozygous mutations in Tousled-like kinase 2 (TLK2) in 38 unrelated individuals and two affected mothers, using whole-exome and whole-genome sequencing technologies, matchmaker databases, and international collaborations. Affected individuals had a consistent phenotype, characterized by mild-borderline neurodevelopmental delay (86%), behavioral disorders (68%), severe gastro-intestinal problems (63%), and facial dysmorphism including blepharophimosis (82%), telecanthus (74%), prominent nasal bridge (68%), broad nasal tip (66%), thin vermilion of the upper lip (62%), and upslanting palpebral fissures (55%). Analysis of cell lines from three affected individuals showed that mutations act through a loss-of-function mechanism in at least two case subjects. Genotype-phenotype analysis and comparison of computationally modeled faces showed that phenotypes of these and other individuals with loss-of-function variants significantly overlapped with phenotypes of individuals with other variant types (missense and C-terminal truncating). This suggests that haploinsufficiency of TLK2 is the most likely underlying disease mechanism, leading to a consistent neurodevelopmental phenotype. This work illustrates the power of international data sharing, by the identification of 40 individuals from 26 different centers in 7 different countries, allowing the identification, clinical delineation, and genotype-phenotype evaluation of a distinct NDD caused by mutations in TLK2.


Asunto(s)
Estudios de Asociación Genética , Patrón de Herencia/genética , Mutación con Pérdida de Función/genética , Trastornos del Neurodesarrollo/genética , Proteínas Quinasas/genética , Adolescente , Adulto , Secuencia de Bases , Línea Celular , Niño , Preescolar , Facies , Femenino , Humanos , Lactante , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Translocación Genética , Adulto Joven
16.
Hum Mol Genet ; 27(11): 1927-1940, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29635513

RESUMEN

Mutations in SNX14 cause the autosomal recessive cerebellar ataxia 20 (SCAR20). Mutations generally result in loss of protein although several coding region deletions have also been reported. Patient-derived fibroblasts show disrupted autophagy, but the precise function of SNX14 is unknown. The yeast homolog, Mdm1, functions in endoplasmic reticulum (ER)-lysosome/vacuole inter-organelle tethering, but functional conservation in mammals is still required. Here, we show that loss of SNX14 alters but does not block autophagic flux. In addition, we find that SNX14 is an ER-associated protein that functions in neutral lipid homeostasis and inter-organelle crosstalk. SNX14 requires its N-terminal transmembrane helices for ER localization, while the Phox homology (PX) domain is dispensable for subcellular localization. Both SNX14-mutant fibroblasts and SNX14KO HEK293 cells accumulate aberrant cytoplasmic vacuoles, suggesting defects in endolysosomal homeostasis. However, ER-late endosome/lysosome contact sites are maintained in SNX14KO cells, indicating that it is not a prerequisite for ER-endolysosomal tethering. Further investigation of SNX14- deficiency indicates general defects in neutral lipid metabolism. SNX14KO cells display distinct perinuclear accumulation of filipin in LAMP1-positive lysosomal structures indicating cholesterol accumulation. Consistent with this, SNX14KO cells display a slight but detectable decrease in cholesterol ester levels, which is exacerbated with U18666A. Finally, SNX14 associates with ER-derived lipid droplets (LD) following oleate treatment, indicating a role in ER-LD crosstalk. We therefore identify an important role for SNX14 in neutral lipid homeostasis between the ER, lysosomes and LDs that may provide an early intervention target to alleviate the clinical symptoms of SCAR20.


Asunto(s)
Retículo Endoplásmico/genética , Metabolismo de los Lípidos/genética , Nexinas de Clasificación/genética , Ataxias Espinocerebelosas/genética , Autofagia/genética , Retículo Endoplásmico/metabolismo , Endosomas , Técnicas de Inactivación de Genes , Células HEK293 , Homeostasis/efectos de los fármacos , Humanos , Proteínas de Filamentos Intermediarios/genética , Gotas Lipídicas/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/genética , Mutación , Ácido Oléico/farmacología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Nexinas de Clasificación/deficiencia , Nexinas de Clasificación/metabolismo , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/fisiopatología
18.
Hum Genet ; 137(2): 111-127, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29305691

RESUMEN

Cerebellar ataxia, areflexia, pes cavus, optic atrophy and sensorineural hearing impairment (CAPOS) is a rare clinically distinct syndrome caused by a single dominant missense mutation, c.2452G>A, p.Glu818Lys, in ATP1A3, encoding the neuron-specific alpha subunit of the Na+/K+-ATPase α3. Allelic mutations cause the neurological diseases rapid dystonia Parkinsonism and alternating hemiplegia of childhood, disorders which do not encompass hearing or visual impairment. We present detailed clinical phenotypic information in 18 genetically confirmed patients from 11 families (10 previously unreported) from Denmark, Sweden, UK and Germany indicating a specific type of hearing impairment-auditory neuropathy (AN). All patients were clinically suspected of CAPOS and had hearing problems. In this retrospective analysis of audiological data, we show for the first time that cochlear outer hair cell activity was preserved as shown by the presence of otoacoustic emissions and cochlear microphonic potentials, but the auditory brainstem responses were grossly abnormal, likely reflecting neural dyssynchrony. Poor speech perception was observed, especially in noise, which was beyond the hearing level obtained in the pure tone audiograms in several of the patients presented here. Molecular modelling and in vitro electrophysiological studies of the specific CAPOS mutation were performed. Heterologous expression studies of α3 with the p.Glu818Lys mutation affects sodium binding to, and release from, the sodium-specific site in the pump, the third ion-binding site. Molecular dynamics simulations confirm that the structure of the C-terminal region is affected. In conclusion, we demonstrate for the first time evidence for auditory neuropathy in CAPOS syndrome, which may reflect impaired propagation of electrical impulses along the spiral ganglion neurons. This has implications for diagnosis and patient management. Auditory neuropathy is difficult to treat with conventional hearing aids, but preliminary improvement in speech perception in some patients suggests that cochlear implantation may be effective in CAPOS patients.


Asunto(s)
Ataxia Cerebelosa/genética , Deformidades Congénitas del Pie/genética , Pérdida Auditiva Central/genética , Pérdida Auditiva Sensorineural/genética , Atrofia Óptica/genética , Reflejo Anormal/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , Adolescente , Adulto , Ataxia Cerebelosa/epidemiología , Ataxia Cerebelosa/fisiopatología , Niño , Preescolar , Dinamarca/epidemiología , Femenino , Deformidades Congénitas del Pie/epidemiología , Deformidades Congénitas del Pie/fisiopatología , Alemania/epidemiología , Pérdida Auditiva Central/epidemiología , Pérdida Auditiva Central/fisiopatología , Pérdida Auditiva Sensorineural/epidemiología , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Masculino , Simulación de Dinámica Molecular , Mutación Missense/genética , Atrofia Óptica/epidemiología , Atrofia Óptica/fisiopatología , Fenotipo , Estudios Retrospectivos , ATPasa Intercambiadora de Sodio-Potasio/química , Suecia/epidemiología , Adulto Joven
19.
Orphanet J Rare Dis ; 12(1): 24, 2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-28173822

RESUMEN

BACKGROUND: We describe molecular diagnosis in a complex consanguineous family: four offspring presented with combinations of three distinctive phenotypes; non-syndromic hearing loss (NSHL), an unusual skeletal phenotype comprising multiple fractures, cranial abnormalities and diaphyseal expansion, and significant developmental delay with microcephaly. We performed Chromosomal Microarray Analysis on the offspring with either the skeletal or developmental delay phenotypes, and linkage analysis and whole exome sequencing (WES) on all four children, parents and maternal aunt. RESULTS: Chromosomal microarray and FISH analysis identified a de novo unbalanced translocation as a cause of the microcephaly and severe developmental delay. WES identified a NSHL-causing splice variant in an autosomal recessive deafness gene PDZD7 which resided in a linkage region and affected three of the children. In the two children diagnosed with an unusual skeletal phenotype, WES eventually disclosed a heterozygous COL1A1 variant which affects C-propetide cleavage site of COL1. The variant was inherited from an apparently unaffected mosaic father in an autosomal dominant fashion. After the discovery of the COL1A1 variant, the skeletal phenotype was diagnosed as a high bone mass form of osteogenesis imperfecta. CONCLUSIONS: Next generation sequencing offers an unbiased approach to molecular genetic diagnosis in highly heterogeneous and poorly characterised disorders and enables early diagnosis as well as detection of mosaicism.


Asunto(s)
Genómica , Pérdida Auditiva/genética , Enfermedades Raras/genética , Preescolar , Consanguinidad , Discapacidades del Desarrollo , Regulación de la Expresión Génica , Humanos , Mutación , Hermanos , Secuenciación del Exoma
20.
Nat Rev Dis Primers ; 3: 16094, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28079113

RESUMEN

Congenital hearing loss (hearing loss that is present at birth) is one of the most prevalent chronic conditions in children. In the majority of developed countries, neonatal hearing screening programmes enable early detection; early intervention will prevent delays in speech and language development and has long-lasting beneficial effects on social and emotional development and quality of life. A diagnosis of hearing loss is usually followed by a search for an underlying aetiology. Congenital hearing loss might be attributed to environmental and prenatal factors, which prevail in low-income settings; congenital infections, particularly cytomegalovirus infection, are also a common risk factor for hearing loss. Genetic causes probably account for the majority of cases in developed countries; mutations can affect any component of the hearing pathway, in particular, inner ear homeostasis (endolymph production and maintenance) and mechano-electrical transduction (the conversion of a mechanical stimulus into electrochemical activity). Once the underlying cause of hearing loss is established, it might direct therapeutic decision making and guide prevention and (genetic) counselling. Management options include specific antimicrobial therapies, surgical treatment of craniofacial abnormalities and implantable or non-implantable hearing devices. An improved understanding of the pathophysiology and molecular mechanisms that underlie hearing loss and increased awareness of recent advances in genetic testing will promote the development of new treatment and screening strategies.


Asunto(s)
Pérdida Auditiva/congénito , Animales , Pérdida Auditiva/epidemiología , Pérdida Auditiva/fisiopatología , Pérdida Auditiva/virología , Humanos , Recién Nacido , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...