Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 109(5): 928-943, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35397207

RESUMEN

Organ fibrosis is a shared endpoint of many diseases, yet underlying mechanisms are not well understood. Several pathways governed by the primary cilium, a sensory antenna present on most vertebrate cells, have been linked with fibrosis. Ciliopathies usually start early in life and represent a considerable disease burden. We performed massively parallel sequencing by using cohorts of genetically unsolved individuals with unexplained liver and kidney failure and correlated this with clinical, imaging, and histopathological analyses. Mechanistic studies were conducted with a vertebrate model and primary cells. We detected bi-allelic deleterious variants in TULP3, encoding a critical adaptor protein for ciliary trafficking, in a total of 15 mostly adult individuals, originating from eight unrelated families, with progressive degenerative liver fibrosis, fibrocystic kidney disease, and hypertrophic cardiomyopathy with atypical fibrotic patterns on histopathology. We recapitulated the human phenotype in adult zebrafish and confirmed disruption of critical ciliary cargo composition in several primary cell lines derived from affected individuals. Further, we show interaction between TULP3 and the nuclear deacetylase SIRT1, with roles in DNA damage repair and fibrosis, and report increased DNA damage ex vivo. Transcriptomic studies demonstrated upregulation of profibrotic pathways with gene clusters for hypertrophic cardiomyopathy and WNT and TGF-ß signaling. These findings identify variants in TULP3 as a monogenic cause for progressive degenerative disease of major organs in which affected individuals benefit from early detection and improved clinical management. Elucidation of mechanisms crucial for DNA damage repair and tissue maintenance will guide novel therapeutic avenues for this and similar genetic and non-genomic diseases.


Asunto(s)
Cardiomiopatía Hipertrófica , Cilios , Adulto , Animales , Cardiomiopatía Hipertrófica/metabolismo , Niño , Cilios/genética , Cilios/metabolismo , Fibrosis , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Riñón , Hígado , Mutación/genética , Pez Cebra/genética
2.
Hum Mol Genet ; 31(14): 2295-2306, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35137054

RESUMEN

Mutations in genes that lead to dysfunctional cilia can cause a broad spectrum of human disease phenotypes referred to as ciliopathies. Many ciliopathy-associated proteins are localized to the evolutionary conserved ciliary transition zone (TZ) subdomain. We identified biallelic missense and nonsense mutations in the gene encoding the transmembrane protein TMEM218 in unrelated patients with features related to Bardet-Biedl, Joubert and Meckel-Gruber syndrome (MKS) and characterized TMEM218 as a major component of the ciliary TZ module. Co-immunoprecipitation assays resulted in the physical interaction of TMEM218 with the MKS module member TMEM67/Meckelin that was significantly reduced by the TMEM218 missense change harboured by one of our patients. We could further validate its pathogenicity by functional in vivo analysis in zebrafish (Danio rerio) as a well-established vertebrate model for ciliopathies. Notably, ciliopathy-related phenotypes were most prominent by genetic interactions with the NPHP module component Nphp4. Conclusively, we describe TMEM218 as a new disease gene for patients with a wide spectrum of syndromic ciliopathy phenotypes and provide evidence for a synergistic interaction of TMEM218 and the NPHP module crucial for proper ciliary function.


Asunto(s)
Anomalías Múltiples , Ciliopatías , Enfermedades Renales Poliquísticas , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Animales , Caenorhabditis elegans/genética , Cilios/genética , Cilios/metabolismo , Trastornos de la Motilidad Ciliar , Ciliopatías/genética , Ciliopatías/metabolismo , Encefalocele , Humanos , Mutación , Enfermedades Renales Poliquísticas/genética , Retinitis Pigmentosa , Pez Cebra/genética
3.
Cell ; 139(6): 1130-42, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20005806

RESUMEN

In mammals, the transcription factor SRY, encoded by the Y chromosome, is normally responsible for triggering the indifferent gonads to develop as testes rather than ovaries. However, testis differentiation can occur in its absence. Here we demonstrate in the mouse that a single factor, the forkhead transcriptional regulator FOXL2, is required to prevent transdifferentiation of an adult ovary to a testis. Inducible deletion of Foxl2 in adult ovarian follicles leads to immediate upregulation of testis-specific genes including the critical SRY target gene Sox9. Concordantly, reprogramming of granulosa and theca cell lineages into Sertoli-like and Leydig-like cell lineages occurs with testosterone levels comparable to those of normal XY male littermates. Our results show that maintenance of the ovarian phenotype is an active process throughout life. They might also have important medical implications for the understanding and treatment of some disorders of sexual development in children and premature menopause in women.


Asunto(s)
Transdiferenciación Celular , Factores de Transcripción Forkhead/metabolismo , Ovario/metabolismo , Testículo/metabolismo , Animales , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/genética , Eliminación de Gen , Células de la Granulosa/citología , Masculino , Ratones , Oocitos/metabolismo , Ovario/citología , Células de Sertoli/citología , Testículo/citología
4.
Genet Med ; 20(6): 614-621, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29309402

RESUMEN

PurposeHearing loss is genetically extremely heterogeneous, making it suitable for next-generation sequencing (NGS). We identified a four-generation family with nonsyndromic mild to severe hearing loss of the mid- to high frequencies and onset from early childhood to second decade in seven members.MethodsNGS of 66 deafness genes, Sanger sequencing, genome-wide linkage analysis, whole-exome sequencing (WES), semiquantitative reverse-transcriptase polymerase chain reaction.ResultsWe identified a heterozygous nonsense mutation, c.6881G>A (p.Trp2294*), in the last coding exon of PTPRQ. PTPRQ has been linked with recessive (DFNB84A), but not dominant deafness. NGS and Sanger sequencing of all exons (including alternatively spliced 5' and N-scan-predicted exons of a putative "extended" transcript) did not identify a second mutation. The highest logarithm of the odds score was in the PTPRQ-containing region on chromosome 12, and p.Trp2294* cosegregated with hearing loss. WES did not identify other cosegregating candidate variants from the mapped region. PTPRQ expression in patient fibroblasts indicated that the mutant allele escapes nonsense-mediated decay (NMD).ConclusionKnown PTPRQ mutations are recessive and do not affect the C-terminal exon. In contrast to recessive loss-of-function mutations, c.6881G>A transcripts may escape NMD. PTPRQTrp2294* protein would lack only six terminal residues and could exert a dominant-negative effect, a possible explanation for allelic deafness, DFNA73, clinically and genetically distinct from DFNB84A.


Asunto(s)
Sordera/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Secuencia de Aminoácidos , Codón sin Sentido/genética , Exoma/genética , Exones/genética , Familia , Femenino , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Pérdida Auditiva/genética , Humanos , Masculino , Mutación , Linaje , Secuenciación del Exoma
5.
Hum Mol Genet ; 22(11): 2177-85, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23418306

RESUMEN

Mutations affecting the integrity and function of cilia have been identified in various genes over the last decade accounting for a group of diseases called ciliopathies. Ciliopathies display a broad spectrum of phenotypes ranging from mild manifestations to lethal combinations of multiple severe symptoms and most of them share cystic kidneys as a common feature. Our starting point was a consanguineous pedigree with three affected fetuses showing an early embryonic phenotype with enlarged cystic kidneys, liver and pancreas and developmental heart disease. By genome-wide linkage analysis, we mapped the disease locus to chromosome 17q11 and identified a homozygous nonsense mutation in NEK8/NPHP9 that encodes a kinase involved in ciliary dynamics and cell cycle progression. Missense mutations in NEK8/NPHP9 have been identified in juvenile cystic kidney jck mice and in patients suffering from nephronophthisis (NPH), an autosomal-recessive cystic kidney disease. This work confirmed a complete loss of NEK8 expression in the affected fetuses due to nonsense-mediated decay. In cultured fibroblasts derived from these fetuses, the expression of prominent polycystic kidney disease genes (PKD1 and PKD2) was decreased, whereas the oncogene c-MYC was upregulated, providing potential explanations for the observed renal phenotype. We furthermore linked NEK8 with NPHP3, another NPH protein known to cause a very similar phenotype in case of null mutations. Both proteins interact and activate the Hippo effector TAZ. Taken together, our study demonstrates that NEK8 is essential for organ development and that the complete loss of NEK8 perturbs multiple signalling pathways resulting in a severe early embryonic phenotype.


Asunto(s)
Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Síndrome de Dandy-Walker/genética , Síndrome de Dandy-Walker/metabolismo , Regulación de la Expresión Génica , Mutación , Quiste Pancreático/genética , Quiste Pancreático/metabolismo , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal , Anomalías Múltiples/patología , Línea Celular , Consanguinidad , Síndrome de Dandy-Walker/patología , Femenino , Feto/anomalías , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Genotipo , Vía de Señalización Hippo , Humanos , Masculino , Quinasas Relacionadas con NIMA , Quiste Pancreático/patología , Linaje , Polimorfismo de Nucleótido Simple , Unión Proteica , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
6.
Hum Mutat ; 35(5): 565-70, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24616153

RESUMEN

MYO1A is considered the gene underlying autosomal dominant nonsyndromic hearing loss DFNA48, based on six missense variants, one small in-frame insertion, and one nonsense mutation. Results from NGS targeting 66 deafness genes in 109 patients identified three families challenging this assumption: two novel nonsense (p.Tyr740* and p.Arg262*) and a known missense variant were identified heterozygously not only in index patients, but also in unaffected relatives. Deafness in these families clearly resulted from mutations in other genes (MYO7A, EYA1, and CIB2). Most of the altogether 10 MYO1A mutations are annotated in dbSNP, and population frequencies (dbSNP, 1000 Genomes, Exome Sequencing Project) above 0.1% contradict pathogenicity under a dominant model. One healthy individual was even homozygous for p.Arg262*, compatible with homozygous Myo1a knockout mice lacking any overt pathology. MYO1A seems dispensable for hearing and overall nonessential. MYO1A adds to the list of "erroneous disease genes", which will expand with increasing availability of large-scale sequencing data.


Asunto(s)
Predisposición Genética a la Enfermedad , Pérdida Auditiva Sensorineural/genética , Mutación Missense , Cadenas Pesadas de Miosina/genética , Miosina Tipo I/genética , Adolescente , Adulto , Animales , Niño , Preescolar , Bases de Datos Genéticas , Femenino , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/patología , Humanos , Lactante , Ratones , Ratones Noqueados , Persona de Mediana Edad , Linaje , Polimorfismo de Nucleótido Simple
7.
Hum Mutat ; 35(10): 1153-62, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25044745

RESUMEN

We describe a consanguineous Iraqi family with Leber congenital amaurosis (LCA), Joubert syndrome (JBTS), and polycystic kidney disease (PKD). Targeted next-generation sequencing for excluding mutations in known LCA and JBTS genes, homozygosity mapping, and whole-exome sequencing identified a homozygous missense variant, c.317G>C (p.Arg106Pro), in POC1B, a gene essential for ciliogenesis, basal body, and centrosome integrity. In silico modeling suggested a requirement of p.Arg106 for the formation of the third WD40 repeat and a protein interaction interface. In human and mouse retina, POC1B localized to the basal body and centriole adjacent to the connecting cilium of photoreceptors and in synapses of the outer plexiform layer. Knockdown of Poc1b in zebrafish caused cystic kidneys and retinal degeneration with shortened and reduced photoreceptor connecting cilia, compatible with the human syndromic ciliopathy. A recent study describes homozygosity for p.Arg106ProPOC1B in a family with nonsyndromic cone-rod dystrophy. The phenotype associated with homozygous p.Arg106ProPOC1B may thus be highly variable, analogous to homozygous p.Leu710Ser in WDR19 causing either isolated retinitis pigmentosa or Jeune syndrome. Our study indicates that POC1B is required for retinal integrity, and we propose POC1B mutations as a probable cause for JBTS with severe PKD.


Asunto(s)
Proteínas de Ciclo Celular/genética , Enfermedades Cerebelosas/genética , Anomalías del Ojo/genética , Enfermedades Renales Quísticas/genética , Mutación , Retina/anomalías , Anomalías Múltiples , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/metabolismo , Enfermedades Cerebelosas/metabolismo , Enfermedades Cerebelosas/patología , Cerebelo/anomalías , Niño , Cilios/metabolismo , Cilios/ultraestructura , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Técnicas de Silenciamiento del Gen , Humanos , Irak , Riñón/patología , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/metabolismo , Masculino , Ratones , Datos de Secuencia Molecular , Linaje , Retina/metabolismo , Retina/patología , Pez Cebra
8.
Pediatr Nephrol ; 29(8): 1451-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24504730

RESUMEN

BACKGROUND: An emerging number of clinically and genetically heterogeneous diseases now collectively termed ciliopathies have been connected to the dysfunction of primary cilia. We describe an 8-year-old girl with a complex phenotype that did not clearly match any familiar syndrome. CASE-DIAGNOSIS/TREATMENT: Hypotonia, facial dysmorphism and retardation were noted shortly after birth. Other features included short stature, mild skeletal anomalies, strabism, deafness, subdural hygroma, hepatosplenomegaly and end-stage renal failure. Renal biopsy revealed tubular atrophy, interstitial fibrosis and segmental glomerulosclerosis. After exclusion of a chromosomal abnormality by array-comparative genomic hybridization (CGH), we performed next-generation sequencing (NGS) using a customized panel that targeted 131 genes known or hypothesized to cause ciliopathies. We identified the novel homozygous WDR19 mutation c.1483G > C (p.Gly495Arg) that affects an evolutionarily highly conserved residue in the intraflagellar transport protein IFT144, is absent from databases and is predicted to be pathogenic by all bioinformatic sources used. CONCLUSION: Mutations in WDR19 encoding the intraflagellar transport component IFT144 have recently been described in single families with the clinically overlapping skeletal ciliopathies Jeune and Sensenbrenner syndromes, combined or isolated nephronophthisis (NPHP) and retinitis pigmentosa (RP) (Senior-Loken syndrome). Our patient emphasizes the usefulness and efficiency of a comprehensive NGS panel approach in patients with unclassified ciliopathies. It further suggests that WDR19 mutations can cause a broad spectrum of ciliopathies that extends to Jeune and Sensenbrenner syndromes, RP and renal NPHP-like phenotypes.


Asunto(s)
Cilios/patología , Enfermedades Renales/genética , Proteínas/genética , Niño , Proteínas del Citoesqueleto , Exones/genética , Femenino , Crecimiento/fisiología , Homocigoto , Humanos , Péptidos y Proteínas de Señalización Intracelular , Enfermedades Renales/patología , Mutación/genética , Análisis de Secuencia de ADN
9.
Hum Mutat ; 34(5): 714-24, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23418020

RESUMEN

Ciliopathies are genetically heterogeneous disorders characterized by variable expressivity and overlaps between different disease entities. This is exemplified by the short rib-polydactyly syndromes, Jeune, Sensenbrenner, and Mainzer-Saldino chondrodysplasia syndromes. These three syndromes are frequently caused by mutations in intraflagellar transport (IFT) genes affecting the primary cilia, which play a crucial role in skeletal and chondral development. Here, we identified mutations in IFT140, an IFT complex A gene, in five Jeune asphyxiating thoracic dystrophy (JATD) and two Mainzer-Saldino syndrome (MSS) families, by screening a cohort of 66 JATD/MSS patients using whole exome sequencing and targeted resequencing of a customized ciliopathy gene panel. We also found an enrichment of rare IFT140 alleles in JATD compared with nonciliopathy diseases, implying putative modifier effects for certain alleles. IFT140 patients presented with mild chest narrowing, but all had end-stage renal failure under 13 years of age and retinal dystrophy when examined for ocular dysfunction. This is consistent with the severe cystic phenotype of Ift140 conditional knockout mice, and the higher level of Ift140 expression in kidney and retina compared with the skeleton at E15.5 in the mouse. IFT140 is therefore a major cause of cono-renal syndromes (JATD and MSS). The present study strengthens the rationale for IFT140 screening in skeletal ciliopathy spectrum patients that have kidney disease and/or retinal dystrophy.


Asunto(s)
Transporte Biológico/genética , Cilios/metabolismo , Enfermedades Renales/genética , Mutación , Animales , Ataxia Cerebelosa/genética , Niño , Estudios de Cohortes , Progresión de la Enfermedad , Exoma , Humanos , Enfermedades Renales/patología , Masculino , Ratones , Retinitis Pigmentosa/genética
10.
PLoS One ; 13(12): e0207958, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30543658

RESUMEN

Retinitis pigmentosa (RP) is an inherited degenerative disease causing severe retinal dystrophy and visual impairment mainly with onset in infancy or adolescence. Targeted next-generation sequencing (NGS) has become an efficient tool to encounter the enormous genetic heterogeneity of diverse retinal dystrophies, including RP. To identify disease-causing mutations in unselected, consecutive RP patients, we conducted Sanger sequencing of genes commonly involved in the suspected genetic RP subtype, followed by targeted large-panel NGS if no mutation was identified, or NGS as primary analysis. A high (70%) detection rate of disease-causing mutations was achieved in a large cohort of 116 unrelated patients. About half (48%) of the solved RP cases were explained by mutations in four genes: RPGR, EYS, PRPF31 and USH2A. Overall, 110 different mutations distributed across 30 different genes were detected, and 46 of these mutations were novel. A molecular diagnosis was achieved in the majority (82-100%) of patients if the family history was suggestive for a particular mode of inheritance, but only in 60% in cases of sporadic RP. The diagnostic potential of extensive molecular analysis in a routine setting is also illustrated by the identification of unexpected genotype-phenotype correlations for RP patients with mutations in CRX, CEP290, RPGRIP1, MFSD8. Furthermore, we identified numerous mutations in autosomal dominant (PRPF31, PRPH2, CRX) and X-linked (RPGR) RP genes in patients with sporadic RP. Variants in RP2 and RPGR were also found in female RP patients with apparently sporadic or dominant disease. In summary, this study demonstrates that massively parallel sequencing of all known retinal dystrophy genes is a valuable diagnostic approach for RP patients.


Asunto(s)
Pruebas Genéticas/métodos , Retinitis Pigmentosa/genética , Adolescente , Adulto , Anciano , Estudios Transversales , Análisis Mutacional de ADN/métodos , Femenino , Genes Ligados a X/genética , Estudios de Asociación Genética , Alemania , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Patrón de Herencia/genética , Masculino , Persona de Mediana Edad , Mutación , Retinitis Pigmentosa/diagnóstico por imagen , Estudios Retrospectivos
11.
Sci Rep ; 8(1): 4824, 2018 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-29555955

RESUMEN

Macular and cone/cone-rod dystrophies (MD/CCRD) demonstrate a broad genetic and phenotypic heterogeneity, with retinal alterations solely or predominantly involving the central retina. Targeted next-generation sequencing (NGS) is an efficient diagnostic tool for identifying mutations in patient with retinitis pigmentosa, which shows similar genetic heterogeneity. To detect the genetic causes of disease in patients with MD/CCRD, we implemented a two-tier procedure consisting of Sanger sequencing and targeted NGS including genes associated with clinically overlapping conditions. Disease-causing mutations were identified in 74% of 251 consecutive MD/CCRD patients (33% of the variants were novel). Mutations in ABCA4, PRPH2 and BEST1 accounted for 57% of disease cases. Further mutations were identified in CDHR1, GUCY2D, PROM1, CRX, GUCA1A, CERKL, MT-TL1, KIF11, RP1L1, MERTK, RDH5, CDH3, C1QTNF5, CRB1, JAG1, DRAM2, POC1B, NPHP1 and RPGR. We provide detailed illustrations of rare phenotypes, including autofluorescence and optical coherence tomography imaging. Targeted NGS also identified six potential novel genotype-phenotype correlations for FAM161A, INPP5E, MERTK, FBLN5, SEMA4A and IMPDH1. Clinical reassessment of genetically unsolved patients revealed subgroups with similar retinal phenotype, indicating a common molecular disease cause in each subgroup.


Asunto(s)
Distrofias de Conos y Bastones/genética , Distrofias de Conos y Bastones/patología , Proteínas del Ojo/genética , Marcadores Genéticos , Degeneración Macular/genética , Degeneración Macular/patología , Mutación , Adulto , Anciano , Estudios Transversales , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Persona de Mediana Edad , Linaje , Pronóstico , Estudios Retrospectivos
12.
Mol Genet Genomic Med ; 5(5): 531-552, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28944237

RESUMEN

BACKGROUND: Combined retinal degeneration and sensorineural hearing impairment is mostly due to autosomal recessive Usher syndrome (USH1: congenital deafness, early retinitis pigmentosa (RP); USH2: progressive hearing impairment, RP). METHODS: Sanger sequencing and NGS of 112 genes (Usher syndrome, nonsyndromic deafness, overlapping conditions), MLPA, and array-CGH were conducted in 138 patients clinically diagnosed with Usher syndrome. RESULTS: A molecular diagnosis was achieved in 97% of both USH1 and USH2 patients, with biallelic mutations in 97% (USH1) and 90% (USH2), respectively. Quantitative readout reliably detected CNVs (confirmed by MLPA or array-CGH), qualifying targeted NGS as one tool for detecting point mutations and CNVs. CNVs accounted for 10% of identified USH2A alleles, often in trans to seemingly monoallelic point mutations. We demonstrate PTC124-induced read-through of the common p.Trp3955* nonsense mutation (13% of detected USH2A alleles), a potential therapy target. Usher gene mutations were found in most patients with atypical Usher syndrome, but the diagnosis was adjusted in case of double homozygosity for mutations in OTOA and NR2E3, genes implicated in isolated deafness and RP. Two patients with additional enamel dysplasia had biallelic PEX26 mutations, for the first time linking this gene to Heimler syndrome. CONCLUSION: Targeted NGS not restricted to Usher genes proved beneficial in uncovering conditions mimicking Usher syndrome.

13.
Hum Gene Ther ; 16(4): 533-9, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15871685

RESUMEN

The inhibition of gene expression by RNA interference harbors a high potential for application in the therapy of human diseases. However, while exogenous application of siRNAs efficiently inhibits gene expression, these effects are only transient in mammalian cells. We designed a short hairpin RNA-expression cassette to target the bcr-abl oncogene that was then introduced into the nonviral vector system pEPI-1, which replicates episomally in the absence of selection in the bcr-abl-positive cell line K562. Forty-two days after transfection the bcr-abl- but not the cytokine-dependent growth rate was found to be drastically reduced in K562 cells. Western analysis revealed a more than 90% reduction in the expression of the fusion protein bcr-abl while the expression of the bcr protein remained unaffected. In addition, we show that the level of bcr-abl mRNA was specifically reduced in these cells for more than 90%. These results demonstrate that the vector system pEPI-1 allows specific and efficient long term gene suppression by using a short hairpin RNA transcription unit.


Asunto(s)
Proteínas de Fusión bcr-abl/genética , Vectores Genéticos/genética , Plásmidos/genética , ARN/farmacología , Replicación Viral/genética , Secuencia de Bases , Northern Blotting , Western Blotting , Proliferación Celular , Citocinas/metabolismo , ADN Polimerasa III/genética , Proteínas de Fusión bcr-abl/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/farmacología , Humanos , Células K562 , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , ARN/química , ARN/genética , ARN Catalítico/química
14.
Br J Ophthalmol ; 99(4): 488-92, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25342276

RESUMEN

BACKGROUND: In Arabian children referred with retinal dystrophy, we have observed that a specific biallelic nonsense mutation in the gene encoding tubby-like protein 1 (TULP1, c.901C>T (p.Gln301*)) is recurrent. This makes the mutation and its associated childhood retinopathy particularly interesting for genetic diagnostic and, potentially, gene therapy approaches. We characterise the ophthalmic phenotype associated with recessive p.Gln301* mutation in TULP1 and assess the mutation for single founder effect. METHODS: Retrospective consecutive case series (2011-2014) of 10 Arabian children (8 families) homozygous for the p.Gln301* mutation (detected after next-generation sequencing) and 12 ethnically matched controls. TULP1 haplotypes were constructed by analysis of TULP1 intragenic single nucleotide polymorphisms from next-generation sequencing data and genotyping of gene-flanking polymorphic microsatellite markers. RESULTS: All 10 children (2-8 years old; mean 5.2, median 6) had nystagmus since soon after birth, a grossly normal posterior pole other than arteriolar attenuation, peripheral mottling with apparent evolution to bone spicules, and hyperopia. Rod function was non-recordable while cone function was present (albeit depressed and delayed); however, repeat electroretinogram years later in two children revealed loss of recordable cone function. Autofluorescence showed a hyper-fluorescent ring around the fovea while central optical coherence tomography was within normal limits. A specific haplotype was associated with p.Gln301* and was not present in controls. CONCLUSIONS: The TULP1 allele p.Gln301* represents a founder mutation on the Arabian Peninsula and is associated with a recognisable congenital recessive rod-cone dystrophy phenotype in the homozygous state.


Asunto(s)
Codón sin Sentido , Proteínas del Ojo/genética , Efecto Fundador , Polimorfismo de Nucleótido Simple , Retinitis Pigmentosa/genética , Alelos , Niño , Preescolar , Electrorretinografía , Femenino , Homocigoto , Humanos , Masculino , Nistagmo Patológico/genética , Fenotipo , Retinitis Pigmentosa/congénito , Estudios Retrospectivos , Arabia Saudita , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología
15.
Br J Ophthalmol ; 99(12): 1725-31, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26294103

RESUMEN

BACKGROUND/AIM: We have noted a phenotype of early-onset retinal dystrophy with macular staphyloma but without high myopia. The aim of this study is to report the underlying genetic mutations and the subcellular localisation of the gene product in the retina. METHODS: Retrospective case series (2012-2015); immunohistochemical analyses of mammalian retina for in situ protein localisation. RESULTS: All three probands were first noted to have decreased vision at 3-6 years old which worsened over time. At ages 39, 37 and 12 years old, all had similar retinal findings: dystrophic changes (retinal pigment epithelium mottling, vessel narrowing), macular staphyloma (despite only mild myopia or high hyperopia), and non-recordable electroretinography. All harboured homozygous mutations in C21orf2, a gene recently suggested to be associated with retinal dystrophy but of unknown function. Two had a frameshift deletion c.436_466del (p.Glu146Serfs*6). The third had a missense mutation affecting a highly conserved residue (p.Cys61Tyr) and was short (below the 3rd percentile) and obese (50th percentile for weight despite short stature). Immunohistochemical studies in human, pig and mouse retinas localised C21orf2 protein to the ciliary structures of the photoreceptor cell (the daughter basal body, the centriole adjacent to the basal body, and the connecting cilium). CONCLUSIONS: This retinal dystrophy phenotype is caused by recessive mutations in C21orf2 and can be considered a retinal ciliopathy as C21orf2 encodes a protein that localises to photoreceptor ciliary structures. The short stature and obesity noted in the youngest girl suggest that for some patients biallelic C21orf2 mutations may result in syndromic ciliopathy.


Asunto(s)
Mutación del Sistema de Lectura/genética , Genes Recesivos , Mutación Missense/genética , Cilio Conector de los Fotorreceptores/metabolismo , Proteínas/genética , Retina/patología , Distrofias Retinianas/genética , Adulto , Secuencia de Aminoácidos , Animales , Western Blotting , Niño , Consanguinidad , Proteínas del Citoesqueleto , Dilatación Patológica , Electrorretinografía , Femenino , Humanos , Imagen por Resonancia Magnética , Ratones , Datos de Secuencia Molecular , Linaje , Reacción en Cadena de la Polimerasa , Proteínas/metabolismo , Estudios Retrospectivos , Sus scrofa , Donantes de Tejidos , Tomografía de Coherencia Óptica
16.
PLoS One ; 10(2): e0116680, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25646624

RESUMEN

Renal cysts are clinically and genetically heterogeneous conditions. Autosomal dominant polycystic kidney disease (ADPKD) is the most frequent life-threatening genetic disease and mainly caused by mutations in PKD1. The presence of six PKD1 pseudogenes and tremendous allelic heterogeneity make molecular genetic testing challenging requiring laborious locus-specific amplification. Increasing evidence suggests a major role for PKD1 in early and severe cases of ADPKD and some patients with a recessive form. Furthermore it is becoming obvious that clinical manifestations can be mimicked by mutations in a number of other genes with the necessity for broader genetic testing. We established and validated a sequence capture based NGS testing approach for all genes known for cystic and polycystic kidney disease including PKD1. Thereby, we demonstrate that the applied standard mapping algorithm specifically aligns reads to the PKD1 locus and overcomes the complication of unspecific capture of pseudogenes. Employing careful and experienced assessment of NGS data, the method is shown to be very specific and equally sensitive as established methods. An additional advantage over conventional Sanger sequencing is the detection of copy number variations (CNVs). Sophisticated bioinformatic read simulation increased the high analytical depth of the validation study and further demonstrated the strength of the approach. We further raise some awareness of limitations and pitfalls of common NGS workflows when applied in complex regions like PKD1 demonstrating that quality of NGS needs more than high coverage of the target region. By this, we propose a time- and cost-efficient diagnostic strategy for comprehensive molecular genetic testing of polycystic kidney disease which is highly automatable and will be of particular value when therapeutic options for PKD emerge and genetic testing is needed for larger numbers of patients.


Asunto(s)
Técnicas de Diagnóstico Molecular/métodos , Enfermedades Renales Poliquísticas/diagnóstico , Enfermedades Renales Poliquísticas/genética , Variaciones en el Número de Copia de ADN , Reacciones Falso Negativas , Duplicación de Gen , Sitios Genéticos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Riñón/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Seudogenes/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora
17.
Orphanet J Rare Dis ; 10: 15, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25759012

RESUMEN

BACKGROUND: Early-onset hearing loss is mostly of genetic origin. The complexity of the hearing process is reflected by its extensive genetic heterogeneity, with probably many causative genes remaining to be identified. Here, we aimed at identifying the genetic basis for autosomal dominant non-syndromic hearing loss (ADNSHL) in a large German family. METHODS: A panel of 66 known deafness genes was analyzed for mutations by next-generation sequencing (NGS) in the index patient. We then conducted genome-wide linkage analysis, and whole-exome sequencing was carried out with samples of two patients. Expression of Osbpl2 in the mouse cochlea was determined by immunohistochemistry. Because Osbpl2 has been proposed as a target of miR-96, we investigated homozygous Mir96 mutant mice for its upregulation. RESULTS: Onset of hearing loss in the investigated ADNSHL family is in childhood, initially affecting the high frequencies and progressing to profound deafness in adulthood. However, there is considerable intrafamilial variability. We mapped a novel ADNSHL locus, DFNA67, to chromosome 20q13.2-q13.33, and subsequently identified a co-segregating heterozygous frameshift mutation, c.141_142delTG (p.Arg50Alafs*103), in OSBPL2, encoding a protein known to interact with the DFNA1 protein, DIAPH1. In mice, Osbpl2 was prominently expressed in stereocilia of cochlear outer and inner hair cells. We found no significant Osbpl2 upregulation at the mRNA level in homozygous Mir96 mutant mice. CONCLUSION: The function of OSBPL2 in the hearing process remains to be determined. Our study and the recent description of another frameshift mutation in a Chinese ADNSHL family identify OSBPL2 as a novel gene for progressive deafness.


Asunto(s)
Sordera/genética , Células Ciliadas Auditivas/metabolismo , Receptores de Esteroides/metabolismo , Estereocilios/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Preescolar , Femenino , Regulación de la Expresión Génica , Ligamiento Genético , Humanos , Lactante , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Linaje , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Esteroides/genética , Adulto Joven
18.
Br J Ophthalmol ; 97(6): 760-4, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23505306

RESUMEN

AIM: To characterise the childhood retinal phenotype associated with recessive mutations in retinitis pigmentosa GTPase regulator interacting protein 1 (RPGRIP1), a gene that has been infrequently associated with Leber congenital amaurosis, the most severe form of childhood non-syndromic retinal dystrophy. METHODS: This was a retrospective case series analysis. RESULTS: Nine children (seven families) with homozygous RPGRIP1 mutations were identified. All were noted by their families to have had shaking eyes, variable eye turn and/or poor vision at or soon after birth and to be more comfortable in darkness than daylight. At the age of examination (2-7 years of age) fixation was poor or non-existent with hemeralopia, nystagmus, variable strabismus and often an oculodigital sign (6/9). Electroretinography was non-recordable. The posterior pole was grossly normal with mild vascular attenuation, but one girl did have a subtle abnormal macular reflex associated with decreased autofluorescence. Retinal pigment epithelium changes were seen in the periphery, ranging from mottling to bone spicules, and cycloplaegic refraction was hyperopic (+3 to +10 diopters). Two children were photophobic and two were developmentally delayed. One boy had oesotropia and nystagmus that decreased when hyperopic spectacles were worn. One girl decreased her nystagmus amplitude by adopting a particular gaze preference. CONCLUSIONS: Recessive RPGRIP1 mutations cause a severe cone-rod Leber congenital amaurosis phenotype, often with poor or no fixation and an oculodigital sign. In the first decade of life retinal changes are clinically most evident in the periphery. Despite the typical severity of the phenotype, fixation can improve for some affected children with wear of the associated hyperopic refraction or by a null point that dampens nystagmus. Spectacle correction of high refractive errors should be encouraged.


Asunto(s)
Amaurosis Congénita de Leber/genética , Proteínas/genética , Distrofias Retinianas/genética , Retinitis Pigmentosa/genética , Niño , Preescolar , Proteínas del Citoesqueleto , Anteojos , Femenino , Genes Recesivos , Humanos , Amaurosis Congénita de Leber/patología , Masculino , Oftalmoscopía , Fenotipo , Errores de Refracción/genética , Errores de Refracción/patología , Errores de Refracción/terapia , Distrofias Retinianas/patología , Retinitis Pigmentosa/patología , Estudios Retrospectivos
19.
PLoS One ; 8(11): e78496, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24265693

RESUMEN

Retinitis pigmentosa (RP) and Leber congenital amaurosis (LCA) are major causes of blindness. They result from mutations in many genes which has long hampered comprehensive genetic analysis. Recently, targeted next-generation sequencing (NGS) has proven useful to overcome this limitation. To uncover "hidden mutations" such as copy number variations (CNVs) and mutations in non-coding regions, we extended the use of NGS data by quantitative readout for the exons of 55 RP and LCA genes in 126 patients, and by including non-coding 5' exons. We detected several causative CNVs which were key to the diagnosis in hitherto unsolved constellations, e.g. hemizygous point mutations in consanguineous families, and CNVs complemented apparently monoallelic recessive alleles. Mutations of non-coding exon 1 of EYS revealed its contribution to disease. In view of the high carrier frequency for retinal disease gene mutations in the general population, we considered the overall variant load in each patient to assess if a mutation was causative or reflected accidental carriership in patients with mutations in several genes or with single recessive alleles. For example, truncating mutations in RP1, a gene implicated in both recessive and dominant RP, were causative in biallelic constellations, unrelated to disease when heterozygous on a biallelic mutation background of another gene, or even non-pathogenic if close to the C-terminus. Patients with mutations in several loci were common, but without evidence for di- or oligogenic inheritance. Although the number of targeted genes was low compared to previous studies, the mutation detection rate was highest (70%) which likely results from completeness and depth of coverage, and quantitative data analysis. CNV analysis should routinely be applied in targeted NGS, and mutations in non-coding exons give reason to systematically include 5'-UTRs in disease gene or exome panels. Consideration of all variants is indispensable because even truncating mutations may be misleading.


Asunto(s)
Variaciones en el Número de Copia de ADN , Exones/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Distrofias Retinianas/genética , Análisis de Secuencia de ADN , Adolescente , Adulto , Niño , Preescolar , Femenino , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación , Linaje , Distrofias Retinianas/diagnóstico , Adulto Joven
20.
Nat Genet ; 45(8): 951-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23793029

RESUMEN

Nephronophthisis is an autosomal recessive cystic kidney disease that leads to renal failure in childhood or adolescence. Most NPHP gene products form molecular networks. Here we identify ANKS6 as a new NPHP family member that connects NEK8 (NPHP9) to INVS (NPHP2) and NPHP3. We show that ANKS6 localizes to the proximal cilium and confirm its role in renal development through knockdown experiments in zebrafish and Xenopus laevis. We also identify six families with ANKS6 mutations affected by nephronophthisis, including severe cardiovascular abnormalities, liver fibrosis and situs inversus. The oxygen sensor HIF1AN hydroxylates ANKS6 and INVS and alters the composition of the ANKS6-INVS-NPHP3 module. Knockdown of Hif1an in Xenopus results in a phenotype that resembles loss of other NPHP proteins. Network analyses uncovered additional putative NPHP proteins and placed ANKS6 at the center of this NPHP module, explaining the overlapping disease manifestation caused by mutation in ANKS6, NEK8, INVS or NPHP3.


Asunto(s)
Enfermedades Renales Quísticas/genética , Cinesinas/genética , Proteínas Nucleares/genética , Proteínas Quinasas/genética , Factores de Transcripción/genética , Animales , Cilios/metabolismo , Consanguinidad , Exones , Técnicas de Silenciamiento del Gen , Humanos , Intrones , Enfermedades Renales Quísticas/metabolismo , Cinesinas/metabolismo , Ratones , Mutación , Quinasas Relacionadas con NIMA , Proteínas Nucleares/metabolismo , Fenotipo , Enfermedades Renales Poliquísticas/genética , Unión Proteica , Mapas de Interacción de Proteínas , Proteínas Quinasas/metabolismo , Transporte de Proteínas , Factores de Transcripción/metabolismo , Xenopus/embriología , Xenopus/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA