Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Clin Cancer Res ; 29(14): 2602-2611, 2023 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-36799931

RESUMEN

PURPOSE: A single maintenance course of a PARP inhibitor (PARPi) improves progression-free survival (PFS) in germline BRCA1/2-mutant high-grade serous ovarian cancer (gBRCAm-HGSOC). The feasibility of a second maintenance course of PARPi was unknown. PATIENTS AND METHODS: Phase II trial with two entry points (EP1, EP2). Patients were recruited prior to rechallenge platinum. Patients with relapsed, gBRCAm-HGSOC were enrolled at EP1 if they were PARPi-naïve. Patients enrolled at EP2 had received their first course of olaparib prior to trial entry. EP1 patients were retreated with olaparib after RECIST complete/partial response (CR/PR) to platinum. EP2 patients were retreated with olaparib ± cediranib after RECIST CR/PR/stable disease to platinum and according to the platinum-free interval. Co-primary outcomes were the proportion of patients who received a second course of olaparib and the proportion who received olaparib retreatment for ≥6 months. Functional homologous recombination deficiency (HRD), somatic copy-number alteration (SCNA), and BRCAm reversions were investigated in tumor and liquid biopsies. RESULTS: Twenty-seven patients were treated (EP1 = 17, EP2 = 10), and 19 were evaluable. Twelve patients (63%) received a second course of olaparib and 4 received olaparib retreatment for ≥6 months. Common grade ≥2 adverse events during olaparib retreatment were anemia, nausea, and fatigue. No cases of MDS/AML occurred. Mean duration of olaparib treatment and retreatment differed (12.1 months vs. 4.4 months; P < 0.001). Functional HRD and SCNA did not predict PFS. A BRCA2 reversion mutation was detected in a post-olaparib liquid biopsy. CONCLUSIONS: A second course of olaparib can be safely administered to women with gBRCAm-HGSOC but is only modestly efficacious. See related commentary by Gonzalez-Ochoa and Oza, p. 2563.


Asunto(s)
Antineoplásicos , Cistadenocarcinoma Seroso , Neoplasias Ováricas , Humanos , Femenino , Inhibidores de Poli(ADP-Ribosa) Polimerasas/efectos adversos , Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/genética , Antineoplásicos/uso terapéutico , Ftalazinas/efectos adversos , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/mortalidad
2.
J Med Genet ; 59(4): 393-398, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33879512

RESUMEN

PURPOSE: The increased adoption of genomic strategies in the clinic makes it imperative for diagnostic laboratories to improve the efficiency of variant interpretation. Clinical exome sequencing (CES) is becoming a valuable diagnostic tool, capable of meeting the diagnostic demand imposed by the vast array of different rare monogenic disorders. We have assessed a clinician-led and phenotype-based approach for virtual gene panel generation for analysis of targeted CES in patients with rare disease in a single institution. METHODS: Retrospective survey of 400 consecutive cases presumed by clinicians to have rare monogenic disorders, referred on singleton basis for targeted CES. We evaluated diagnostic yield and variant workload to characterise the usefulness of a clinician-led approach for generation of virtual gene panels that can incorporate up to three different phenotype-driven gene selection methods. RESULTS: Abnormalities of the nervous system (54.5%), including intellectual disability, head and neck (19%), skeletal system (16%), ear (15%) and eye (15%) were the most common clinical features reported in referrals. Combined phenotype-driven strategies for virtual gene panel generation were used in 57% of cases. On average, 7.3 variants (median=5) per case were retained for clinical interpretation. The overall diagnostic rate of proband-only CES using personalised phenotype-driven virtual gene panels was 24%. CONCLUSIONS: Our results show that personalised virtual gene panels are a cost-effective approach for variant analysis of CES, maintaining diagnostic yield and optimising the use of resources for clinical genomic sequencing in the clinic.


Asunto(s)
Exoma , Enfermedades Raras , Exoma/genética , Humanos , Enfermedades Raras/genética , Estudios Retrospectivos , Secuenciación del Exoma , Carga de Trabajo
3.
Am J Hum Genet ; 108(11): 2195-2204, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34715011

RESUMEN

Human mitochondrial RNase P (mt-RNase P) is responsible for 5' end processing of mitochondrial precursor tRNAs, a vital step in mitochondrial RNA maturation, and is comprised of three protein subunits: TRMT10C, SDR5C1 (HSD10), and PRORP. Pathogenic variants in TRMT10C and SDR5C1 are associated with distinct recessive or x-linked infantile onset disorders, resulting from defects in mitochondrial RNA processing. We report four unrelated families with multisystem disease associated with bi-allelic variants in PRORP, the metallonuclease subunit of mt-RNase P. Affected individuals presented with variable phenotypes comprising sensorineural hearing loss, primary ovarian insufficiency, developmental delay, and brain white matter changes. Fibroblasts from affected individuals in two families demonstrated decreased steady state levels of PRORP, an accumulation of unprocessed mitochondrial transcripts, and decreased steady state levels of mitochondrial-encoded proteins, which were rescued by introduction of the wild-type PRORP cDNA. In mt-tRNA processing assays performed with recombinant mt-RNase P proteins, the disease-associated variants resulted in diminished mitochondrial tRNA processing. Identification of disease-causing variants in PRORP indicates that pathogenic variants in all three subunits of mt-RNase P can cause mitochondrial dysfunction, each with distinct pleiotropic clinical presentations.


Asunto(s)
Alelos , Pleiotropía Genética , Mitocondrias/enzimología , ARN Mitocondrial/genética , ARN de Transferencia/genética , Ribonucleasa P/genética , Adulto , Femenino , Humanos , Masculino , Linaje
6.
Clin Genet ; 96(6): 515-520, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31441039

RESUMEN

CHRM3 codes for the M3 muscarinic acetylcholine receptor that is located on the surface of smooth muscle cells of the detrusor, the muscle that effects urinary voiding. Previously, we reported brothers in a family affected by a congenital prune belly-like syndrome with mydriasis due to homozygous CHRM3 frameshift variants. In this study, we describe two sisters with bladders that failed to empty completely and pupils that failed to constrict fully in response to light, who are homozygous for the missense CHRM3 variant c.352G > A; p.(Gly118Arg). Samples were not available for genotyping from their brother, who had a history of multiple urinary tract infections and underwent surgical bladder draining in the first year of life. He died at the age of 6 years. This is the first independent report of biallelic variants in CHRM3 in a family with a rare serious bladder disorder associated with mydriasis and provides important evidence of this association.


Asunto(s)
Mutación Missense/genética , Receptor Muscarínico M3/genética , Enfermedades de la Vejiga Urinaria/genética , Secuencia de Bases , Familia , Femenino , Homocigoto , Humanos , Malasia , Masculino
7.
Circ Res ; 124(4): 553-563, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30582441

RESUMEN

RATIONALE: Familial recurrence studies provide strong evidence for a genetic component to the predisposition to sporadic, nonsyndromic Tetralogy of Fallot (TOF), the most common cyanotic congenital heart disease phenotype. Rare genetic variants have been identified as important contributors to the risk of congenital heart disease, but relatively small numbers of TOF cases have been studied to date. OBJECTIVE: We used whole exome sequencing to assess the prevalence of unique, deleterious variants in the largest cohort of nonsyndromic TOF patients reported to date. METHODS AND RESULTS: Eight hundred twenty-nine TOF patients underwent whole exome sequencing. The presence of unique, deleterious variants was determined; defined by their absence in the Genome Aggregation Database and a scaled combined annotation-dependent depletion score of ≥20. The clustering of variants in 2 genes, NOTCH1 and FLT4, surpassed thresholds for genome-wide significance (assigned as P<5×10-8) after correction for multiple comparisons. NOTCH1 was most frequently found to harbor unique, deleterious variants. Thirty-one changes were observed in 37 probands (4.5%; 95% CI, 3.2%-6.1%) and included 7 loss-of-function variants 22 missense variants and 2 in-frame indels. Sanger sequencing of the unaffected parents of 7 cases identified 5 de novo variants. Three NOTCH1 variants (p.G200R, p.C607Y, and p.N1875S) were subjected to functional evaluation, and 2 showed a reduction in Jagged1-induced NOTCH signaling. FLT4 variants were found in 2.4% (95% CI, 1.6%-3.8%) of TOF patients, with 21 patients harboring 22 unique, deleterious variants. The variants identified were distinct to those that cause the congenital lymphoedema syndrome Milroy disease. In addition to NOTCH1, FLT4 and the well-established TOF gene, TBX1, we identified potential association with variants in several other candidates, including RYR1, ZFPM1, CAMTA2, DLX6, and PCM1. CONCLUSIONS: The NOTCH1 locus is the most frequent site of genetic variants predisposing to nonsyndromic TOF, followed by FLT4. Together, variants in these genes are found in almost 7% of TOF patients.


Asunto(s)
Exoma , Tasa de Mutación , Tetralogía de Fallot/genética , Autoantígenos/genética , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Proteínas de Homeodominio/genética , Humanos , Mutación con Pérdida de Función , Mutación Missense , Proteínas Nucleares/genética , Receptor Notch1/genética , Transactivadores/genética , Factores de Transcripción/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
8.
J Med Genet ; 55(2): 114-121, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29074561

RESUMEN

BACKGROUND: Diagnostic use of gene panel next-generation sequencing (NGS) techniques is commonplace for individuals with inherited retinal dystrophies (IRDs), a highly genetically heterogeneous group of disorders. However, these techniques have often failed to capture the complete spectrum of genomic variation causing IRD, including CNVs. This study assessed the applicability of introducing CNV surveillance into first-tier diagnostic gene panel NGS services for IRD. METHODS: Three read-depth algorithms were applied to gene panel NGS data sets for 550 referred individuals, and informatics strategies used for quality assurance and CNV filtering. CNV events were confirmed and reported to referring clinicians through an accredited diagnostic laboratory. RESULTS: We confirmed the presence of 33 deletions and 11 duplications, determining these findings to contribute to the confirmed or provisional molecular diagnosis of IRD for 25 individuals. We show that at least 7% of individuals referred for diagnostic testing for IRD have a CNV within genes relevant to their clinical diagnosis, and determined a positive predictive value of 79% for the employed CNV filtering techniques. CONCLUSION: Incorporation of CNV analysis increases diagnostic yield of gene panel NGS diagnostic tests for IRD, increases clarity in diagnostic reporting and expands the spectrum of known disease-causing mutations.


Asunto(s)
Variaciones en el Número de Copia de ADN , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Distrofias Retinianas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Algoritmos , Proteínas del Citoesqueleto , Duplicación de Gen , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Proteínas de la Membrana/genética , Ribonucleoproteínas Nucleares Pequeñas/genética , Flujo de Trabajo
9.
Am J Hum Genet ; 101(6): 1021-1033, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29220674

RESUMEN

ACTB encodes ß-actin, an abundant cytoskeletal housekeeping protein. In humans, postulated gain-of-function missense mutations cause Baraitser-Winter syndrome (BRWS), characterized by intellectual disability, cortical malformations, coloboma, sensorineural deafness, and typical facial features. To date, the consequences of loss-of-function ACTB mutations have not been proven conclusively. We describe heterozygous ACTB deletions and nonsense and frameshift mutations in 33 individuals with developmental delay, apparent intellectual disability, increased frequency of internal organ malformations (including those of the heart and the renal tract), growth retardation, and a recognizable facial gestalt (interrupted wavy eyebrows, dense eyelashes, wide nose, wide mouth, and a prominent chin) that is distinct from characteristics of individuals with BRWS. Strikingly, this spectrum overlaps with that of several chromatin-remodeling developmental disorders. In wild-type mouse embryos, ß-actin expression was prominent in the kidney, heart, and brain. ACTB mRNA expression levels in lymphoblastic lines and fibroblasts derived from affected individuals were decreased in comparison to those in control cells. Fibroblasts derived from an affected individual and ACTB siRNA knockdown in wild-type fibroblasts showed altered cell shape and migration, consistent with known roles of cytoplasmic ß-actin. We also demonstrate that ACTB haploinsufficiency leads to reduced cell proliferation, altered expression of cell-cycle genes, and decreased amounts of nuclear, but not cytoplasmic, ß-actin. In conclusion, we show that heterozygous loss-of-function ACTB mutations cause a distinct pleiotropic malformation syndrome with intellectual disability. Our biological studies suggest that a critically reduced amount of this protein alters cell shape, migration, proliferation, and gene expression to the detriment of brain, heart, and kidney development.


Asunto(s)
Anomalías Múltiples/genética , Actinas/genética , Discapacidades del Desarrollo/genética , Haploinsuficiencia/genética , Actinas/biosíntesis , Adolescente , Adulto , Anciano , Animales , Ciclo Celular/genética , Proliferación Celular/genética , Niño , Preescolar , Codón sin Sentido/genética , Coloboma/genética , Facies , Femenino , Mutación del Sistema de Lectura/genética , Eliminación de Gen , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/genética , Masculino , Malformaciones del Desarrollo Cortical/genética , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética , Adulto Joven
10.
Arch Dis Child ; 102(11): 1019-1029, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28468868

RESUMEN

BACKGROUND: Inborn errors of metabolism (IEMs) underlie a substantial proportion of paediatric disease burden but their genetic diagnosis can be challenging using the traditional approaches. METHODS: We designed and validated a next-generation sequencing (NGS) panel of 226 IEM genes, created six overlapping phenotype-based subpanels and tested 102 individuals, who presented clinically with suspected childhood-onset IEMs. RESULTS: In 51/102 individuals, NGS fully or partially established the molecular cause or identified other actionable diagnoses. Causal mutations were identified significantly more frequently when the biochemical phenotype suggested a specific IEM or a group of IEMs (p<0.0001), demonstrating the pivotal role of prior biochemical testing in guiding NGS analysis. The NGS panel helped to avoid further invasive, hazardous, lengthy or expensive investigations in 69% individuals (p<0.0001). Additional functional testing due to novel or unexpected findings had to be undertaken in only 3% of subjects, demonstrating that the use of NGS does not significantly increase the burden of subsequent follow-up testing. Even where a molecular diagnosis could not be achieved, NGS-based approach assisted in the management and counselling by reducing the likelihood of a high-penetrant genetic cause. CONCLUSION: NGS has significant clinical utility for the diagnosis of IEMs. Biochemical testing and NGS analysis play complementary roles in the diagnosis of IEMs. Incorporating NGS into the diagnostic algorithm of IEMs can improve the accuracy of diagnosis.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Errores Innatos del Metabolismo/diagnóstico , Adolescente , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Errores Innatos del Metabolismo/genética , Adulto Joven
11.
Eur J Hum Genet ; 25(6): 719-724, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28378820

RESUMEN

Although a common cause of disease, copy number variants (CNVs) have not routinely been identified from next-generation sequencing (NGS) data in a clinical context. This study aimed to examine the sensitivity and specificity of a widely used software package, ExomeDepth, to identify CNVs from targeted NGS data sets. We benchmarked the accuracy of CNV detection using ExomeDepth v1.1.6 applied to targeted NGS data sets by comparison to CNV events detected through whole-genome sequencing for 25 individuals and determined the sensitivity and specificity of ExomeDepth applied to these targeted NGS data sets to be 100% and 99.8%, respectively. To define quality assurance metrics for CNV surveillance through ExomeDepth, we undertook simulation of single-exon (n=1000) and multiple-exon heterozygous deletion events (n=1749), determining a sensitivity of 97% (n=2749). We identified that the extent of sequencing coverage, the inter- and intra-sample variability in the depth of sequencing coverage and the composition of analysis regions are all important determinants of successful CNV surveillance through ExomeDepth. We then applied these quality assurance metrics during CNV surveillance for 140 individuals across 12 distinct clinical areas, encompassing over 500 potential rare disease diagnoses. All 140 individuals lacked molecular diagnoses after routine clinical NGS testing, and by application of ExomeDepth, we identified 17 CNVs contributing to the cause of a Mendelian disorder. Our findings support the integration of CNV detection using ExomeDepth v1.1.6 with routine targeted NGS diagnostic services for Mendelian disorders. Implementation of this strategy increases diagnostic yields and enhances clinical care.


Asunto(s)
Variaciones en el Número de Copia de ADN , Pruebas Genéticas/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Exoma , Pruebas Genéticas/normas , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Humanos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Análisis de Secuencia de ADN/normas
12.
Am J Med Genet A ; 173(4): 1051-1055, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28328138

RESUMEN

PTRH2 is an evolutionarily highly conserved mitochondrial protein that belongs to a family of peptidyl-tRNA hydrolases. Recently, patients from two consanguineous families with mutations in the PTRH2 gene were reported. Global developmental delay associated with microcephaly, growth retardation, progressive ataxia, distal muscle weakness with ankle contractures, demyelinating sensorimotor neuropathy, and sensorineural hearing loss were present in all patients, while facial dysmorphism with widely spaced eyes, exotropia, thin upper lip, proximally placed thumbs, and deformities of the fingers and toes were present in some individuals. Here, we report a new family with three siblings affected by sensorineural hearing loss and peripheral neuropathy. Autozygosity mapping followed by exome sequencing identified a previously reported homozygous missense mutation in PTRH2 (c.254A>C; p.(Gln85Pro)). Sanger sequencing confirmed that the variant segregated with the phenotype. In contrast to the previously reported patient, the affected siblings had normal intelligence, milder microcephaly, delayed puberty, myopia, and moderate insensitivity to pain. Our findings expand the clinical phenotype and further demonstrate the clinical heterogeneity related to PTRH2 variants.


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Pérdida Auditiva Sensorineural/genética , Homocigoto , Proteínas Mitocondriales/genética , Mutación Missense , Enfermedades del Sistema Nervioso Periférico/genética , Adolescente , Secuencia de Bases , Consanguinidad , Progresión de la Enfermedad , Femenino , Expresión Génica , Heterogeneidad Genética , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Masculino , Miopía/fisiopatología , Insensibilidad Congénita al Dolor/fisiopatología , Linaje , Enfermedades del Sistema Nervioso Periférico/diagnóstico , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Fenotipo , Pubertad Tardía/fisiopatología , Hermanos
14.
Orphanet J Rare Dis ; 11(1): 125, 2016 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-27628848

RESUMEN

BACKGROUND: Although the majority of small in-frame insertions/deletions (indels) has no/little affect on protein function, a small subset of these changes has been causally associated with genetic disorders. Notably, the molecular mechanisms and frequency by which they give rise to disease phenotypes remain largely unknown. The aim of this study is to provide insights into the role of in-frame indels (≤21 nucleotides) in two genetically heterogeneous eye disorders. RESULTS: One hundred eighty-one probands with childhood cataracts and 486 probands with retinal dystrophy underwent multigene panel testing in a clinical diagnostic laboratory. In-frame indels were collected and evaluated both clinically and in silico. Variants that could be modeled in the context of protein structure were identified and analysed using integrative structural modeling. Overall, 55 small in-frame indels were detected in 112 of 667 probands (16.8 %); 17 of these changes were novel to this study and 18 variants were reported clinically. A reliable model of the corresponding protein sequence could be generated for 8 variants. Structural modeling indicated a diverse range of molecular mechanisms of disease including disruption of secondary and tertiary protein structure and alteration of protein-DNA binding sites. CONCLUSIONS: In childhood cataract and retinal dystrophy subjects, one small in-frame indel is clinically reported in every ~37 individuals tested. The clinical utility of computational tools evaluating these changes increases when the full complexity of the involved molecular mechanisms is embraced.


Asunto(s)
Oftalmopatías/genética , Mutación INDEL/genética , Sistemas de Lectura/genética , Catarata/genética , Biología Computacional , Humanos , Distrofias Retinianas/genética
15.
Eur J Med Genet ; 59(11): 577-583, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27667191

RESUMEN

Raine syndrome is a rare autosomal recessive bone dysplasia characterized by characteristic facial features with exophthalmos and generalized osteosclerosis. Amelogenesis imperfecta, hearing loss, seizures, and intracerebral calcification are apparent in some affected individuals. Originally, Raine syndrome was originally reported as a lethal syndrome. However, recently a milder phenotype, compatible with life, has been described. Biallelic variants inFAM20C, encoding aGolgi casein kinase involved in biomineralisation, have been identified in affected individuals. We report here a consanguineous Moroccan family with two affected siblingsa girl aged 18 and a boy of 15years. Clinical features, including learning disability, seizures and amelogenesis imperfecta, initially suggested a diagnosis of Kohlschutter-Tonz syndrome. However,a novel homozygous FAM20Cvariantc.676T > A, p.(Trp226Arg) was identified in the affected siblings. Our report reinforces that Raine syndrome is compatible with life, and that mild hypophosphatemia and amelogenesis imperfecta are key features of the attenuated form.


Asunto(s)
Anomalías Múltiples/genética , Amelogénesis Imperfecta/genética , Quinasa de la Caseína I/genética , Fisura del Paladar/genética , Demencia/genética , Diagnóstico Diferencial , Epilepsia/genética , Exoftalmia/genética , Proteínas de la Matriz Extracelular/genética , Microcefalia/genética , Osteosclerosis/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/mortalidad , Anomalías Múltiples/fisiopatología , Adolescente , Amelogénesis Imperfecta/diagnóstico , Amelogénesis Imperfecta/mortalidad , Amelogénesis Imperfecta/fisiopatología , Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/mortalidad , Enfermedades del Desarrollo Óseo/fisiopatología , Fisura del Paladar/diagnóstico , Fisura del Paladar/mortalidad , Fisura del Paladar/fisiopatología , Demencia/diagnóstico , Demencia/mortalidad , Demencia/fisiopatología , Epilepsia/diagnóstico , Epilepsia/mortalidad , Epilepsia/fisiopatología , Exoftalmia/diagnóstico , Exoftalmia/mortalidad , Exoftalmia/fisiopatología , Femenino , Humanos , Discapacidades para el Aprendizaje/genética , Discapacidades para el Aprendizaje/fisiopatología , Masculino , Microcefalia/diagnóstico , Microcefalia/mortalidad , Microcefalia/fisiopatología , Osteosclerosis/diagnóstico , Osteosclerosis/mortalidad , Osteosclerosis/fisiopatología , Fenotipo , Convulsiones/genética , Convulsiones/fisiopatología
16.
Nat Genet ; 48(10): 1185-92, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27571260

RESUMEN

Although ribosomes are ubiquitous and essential for life, recent data indicate that monogenic causes of ribosomal dysfunction can confer a remarkable degree of specificity in terms of human disease phenotype. Box C/D small nucleolar RNAs (snoRNAs) are evolutionarily conserved non-protein-coding RNAs involved in ribosome biogenesis. Here we show that biallelic mutations in the gene SNORD118, encoding the box C/D snoRNA U8, cause the cerebral microangiopathy leukoencephalopathy with calcifications and cysts (LCC), presenting at any age from early childhood to late adulthood. These mutations affect U8 expression, processing and protein binding and thus implicate U8 as essential in cerebral vascular homeostasis.


Asunto(s)
Enfermedades de los Pequeños Vasos Cerebrales/genética , Leucoencefalopatías/genética , Mutación , ARN Nucleolar Pequeño/genética , Adolescente , Adulto , Calcinosis/genética , Calcinosis/patología , Línea Celular , Enfermedades de los Pequeños Vasos Cerebrales/patología , Niño , Preescolar , Cromosomas Humanos Par 17 , Estudios de Cohortes , Quistes/genética , Quistes/patología , Exoma , Femenino , Ligamiento Genético , Genoma Humano , Humanos , Lactante , Leucoencefalopatías/patología , Masculino , Persona de Mediana Edad , Análisis de Secuencia de ADN , Adulto Joven
17.
J Med Genet ; 53(11): 761-767, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27208204

RESUMEN

BACKGROUND: Inherited retinal diseases (IRDs) are a clinically and genetically heterogeneous set of disorders, for which diagnostic second-generation sequencing (next-generation sequencing, NGS) services have been developed worldwide. METHODS: We present the molecular findings of 537 individuals referred to a 105-gene diagnostic NGS test for IRDs. We assess the diagnostic yield, the spectrum of clinical referrals, the variant analysis burden and the genetic heterogeneity of IRD. We retrospectively analyse disease-causing variants, including an assessment of variant frequency in Exome Aggregation Consortium (ExAC). RESULTS: Individuals were referred from 10 clinically distinct classifications of IRD. Of the 4542 variants clinically analysed, we have reported 402 mutations as a cause or a potential cause of disease in 62 of the 105 genes surveyed. These variants account or likely account for the clinical diagnosis of IRD in 51% of the 537 referred individuals. 144 potentially disease-causing mutations were identified as novel at the time of clinical analysis, and we further demonstrate the segregation of known disease-causing variants among individuals with IRD. We show that clinically analysed variants indicated as rare in dbSNP and the Exome Variant Server remain rare in ExAC, and that genes discovered as a cause of IRD in the post-NGS era are rare causes of IRD in a population of clinically surveyed individuals. CONCLUSIONS: Our findings illustrate the continued powerful utility of custom-gene panel diagnostic NGS tests for IRD in the clinic, but suggest clear future avenues for increasing diagnostic yields.

18.
Ophthalmology ; 123(5): 1143-50, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26872967

RESUMEN

PURPOSE: To compare the efficacy of whole genome sequencing (WGS) with targeted next-generation sequencing (NGS) in the diagnosis of inherited retinal disease (IRD). DESIGN: Case series. PARTICIPANTS: A total of 562 patients diagnosed with IRD. METHODS: We performed a direct comparative analysis of current molecular diagnostics with WGS. We retrospectively reviewed the findings from a diagnostic NGS DNA test for 562 patients with IRD. A subset of 46 of 562 patients (encompassing potential clinical outcomes of diagnostic analysis) also underwent WGS, and we compared mutation detection rates and molecular diagnostic yields. In addition, we compared the sensitivity and specificity of the 2 techniques to identify known single nucleotide variants (SNVs) using 6 control samples with publically available genotype data. MAIN OUTCOME MEASURES: Diagnostic yield of genomic testing. RESULTS: Across known disease-causing genes, targeted NGS and WGS achieved similar levels of sensitivity and specificity for SNV detection. However, WGS also identified 14 clinically relevant genetic variants through WGS that had not been identified by NGS diagnostic testing for the 46 individuals with IRD. These variants included large deletions and variants in noncoding regions of the genome. Identification of these variants confirmed a molecular diagnosis of IRD for 11 of the 33 individuals referred for WGS who had not obtained a molecular diagnosis through targeted NGS testing. Weighted estimates, accounting for population structure, suggest that WGS methods could result in an overall 29% (95% confidence interval, 15-45) uplift in diagnostic yield. CONCLUSIONS: We show that WGS methods can detect disease-causing genetic variants missed by current NGS diagnostic methodologies for IRD and thereby demonstrate the clinical utility and additional value of WGS.


Asunto(s)
Enfermedades Hereditarias del Ojo/genética , Genoma , Técnicas de Diagnóstico Molecular , Enfermedades de la Retina/genética , Análisis de Secuencia de ADN , Femenino , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Estudios Retrospectivos , Sensibilidad y Especificidad
19.
Am J Med Genet A ; 170A(5): 1216-24, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26789649

RESUMEN

The 3MC syndromes are a group of rare autosomal recessive disorders where the main clinical features are cleft lip and palate, hypertelorism, highly arched eyebrows, caudal appendage, postnatal growth deficiency, and genitourinary tract anomalies. Ophthalmological abnormalities, most notably anterior chamber defects may also be seen. We describe the clinical and molecular findings in 13 individuals with suspected 3MC syndrome from 12 previously unreported families. The exclusion of the MASP1 and COLEC11 Loci in two individuals from different consanguineous families and the absence of mutations in four further individuals sequenced for both genes raises the possibility that that there is further genetic heterogeneity of 3MC syndrome.


Asunto(s)
Labio Leporino/genética , Fisura del Paladar/genética , Colectinas/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Adolescente , Niño , Preescolar , Labio Leporino/fisiopatología , Fisura del Paladar/fisiopatología , Anomalías del Ojo/genética , Anomalías del Ojo/fisiopatología , Cara/anomalías , Cara/fisiopatología , Femenino , Humanos , Hipertelorismo/genética , Hipertelorismo/fisiopatología , Lactante , Masculino , Mutación , Análisis de Secuencia , Anomalías Urogenitales/genética , Anomalías Urogenitales/fisiopatología
20.
J Hum Genet ; 60(12): 781-5, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26377242

RESUMEN

Dubowitz syndrome is a presumed autosomal recessive disorder characterized by multiple congenital abnormalities: microcephaly, learning and developmental delay, growth failure, and a predisposition to allergies and eczema. There have been more than 150 individuals reported to have this diagnosis, but no unifying genetic alteration has been identified indicating genetic heterogeneity. We report on a pair of monozygotic twins diagnosed clinically with Dubowitz syndrome by Professor Dubowitz over 30 years ago and identified to have a de novo heterozygous 3.2-Mb deletion at 19q13.11q13.12. Exome sequencing did not identify either a putative pathogenic variant on the trans allele supporting recessive inheritance or any other causative sequence variants. Comparison of the phenotype in our cases shows considerable overlap with the 19q13.11 microdeletion syndrome, suggesting that a subset of individuals diagnosed with Dubowitz syndrome may be due to deletions at 19q13. Our finding further reinforces the genetic and phenotypic heterogeneity of Dubowitz syndrome.


Asunto(s)
Alelos , Secuencia de Bases , Cromosomas Humanos Par 19/genética , Eccema/genética , Trastornos del Crecimiento/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Eliminación de Secuencia , Gemelos Monocigóticos/genética , Adulto , Eccema/patología , Facies , Trastornos del Crecimiento/patología , Humanos , Discapacidad Intelectual/patología , Masculino , Microcefalia/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...