Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 20(5): e1011230, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38713708

RESUMEN

Fuchs endothelial corneal dystrophy (FECD) is an age-related cause of vision loss, and the most common repeat expansion-mediated disease in humans characterised to date. Up to 80% of European FECD cases have been attributed to expansion of a non-coding CTG repeat element (termed CTG18.1) located within the ubiquitously expressed transcription factor encoding gene, TCF4. The non-coding nature of the repeat and the transcriptomic complexity of TCF4 have made it extremely challenging to experimentally decipher the molecular mechanisms underlying this disease. Here we comprehensively describe CTG18.1 expansion-driven molecular components of disease within primary patient-derived corneal endothelial cells (CECs), generated from a large cohort of individuals with CTG18.1-expanded (Exp+) and CTG 18.1-independent (Exp-) FECD. We employ long-read, short-read, and spatial transcriptomic techniques to interrogate expansion-specific transcriptomic biomarkers. Interrogation of long-read sequencing and alternative splicing analysis of short-read transcriptomic data together reveals the global extent of altered splicing occurring within Exp+ FECD, and unique transcripts associated with CTG18.1-expansions. Similarly, differential gene expression analysis highlights the total transcriptomic consequences of Exp+ FECD within CECs. Furthermore, differential exon usage, pathway enrichment and spatial transcriptomics reveal TCF4 isoform ratio skewing solely in Exp+ FECD with potential downstream functional consequences. Lastly, exome data from 134 Exp- FECD cases identified rare (minor allele frequency <0.005) and potentially deleterious (CADD>15) TCF4 variants in 7/134 FECD Exp- cases, suggesting that TCF4 variants independent of CTG18.1 may increase FECD risk. In summary, our study supports the hypothesis that at least two distinct pathogenic mechanisms, RNA toxicity and TCF4 isoform-specific dysregulation, both underpin the pathophysiology of FECD. We anticipate these data will inform and guide the development of translational interventions for this common triplet-repeat mediated disease.


Asunto(s)
Distrofia Endotelial de Fuchs , Factor de Transcripción 4 , Expansión de Repetición de Trinucleótido , Humanos , Masculino , Empalme Alternativo/genética , Células Endoteliales/metabolismo , Endotelio Corneal/metabolismo , Endotelio Corneal/patología , Distrofia Endotelial de Fuchs/genética , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Transcriptoma/genética , Expansión de Repetición de Trinucleótido/genética , Femenino
2.
J Med Genet ; 60(4): 368-379, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35882526

RESUMEN

BACKGROUND: Axenfeld-Rieger syndrome (ARS) is characterised by typical anterior segment anomalies, with or without systemic features. The discovery of causative genes identified ARS subtypes with distinct phenotypes, but our understanding is incomplete, complicated by the rarity of the condition. METHODS: Genetic and phenotypic characterisation of the largest reported ARS cohort through comprehensive genetic and clinical data analyses. RESULTS: 128 individuals with causative variants in PITX2 or FOXC1, including 81 new cases, were investigated. Ocular anomalies showed significant overlap but with broader variability and earlier onset of glaucoma for FOXC1-related ARS. Systemic anomalies were seen in all individuals with PITX2-related ARS and the majority of those with FOXC1-related ARS. PITX2-related ARS demonstrated typical umbilical anomalies and dental microdontia/hypodontia/oligodontia, along with a novel high rate of Meckel diverticulum. FOXC1-related ARS exhibited characteristic hearing loss and congenital heart defects as well as previously unrecognised phenotypes of dental enamel hypoplasia and/or crowding, a range of skeletal and joint anomalies, hypotonia/early delay and feeding disorders with structural oesophageal anomalies in some. Brain imaging revealed highly penetrant white matter hyperintensities, colpocephaly/ventriculomegaly and frequent arachnoid cysts. The expanded phenotype of FOXC1-related ARS identified here was found to fully overlap features of De Hauwere syndrome. The results were used to generate gene-specific management plans for the two types of ARS. CONCLUSION: Since clinical features of ARS vary significantly based on the affected gene, it is critical that families are provided with a gene-specific diagnosis, PITX2-related ARS or FOXC1-related ARS. De Hauwere syndrome is proposed to be a FOXC1opathy.


Asunto(s)
Anomalías del Ojo , Proteínas de Homeodominio , Humanos , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Segmento Anterior del Ojo/anomalías , Anomalías del Ojo/genética , Anomalías del Ojo/diagnóstico , Factores de Transcripción Forkhead/genética , Mutación
3.
Int J Mol Sci ; 25(7)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38612555

RESUMEN

We aim to report the ocular phenotype and molecular genetic findings in two Czech families with Sorsby fundus dystrophy and to review all the reported TIMP3 pathogenic variants. Two probands with Sorsby fundus dystrophy and three first-degree relatives underwent ocular examination and retinal imaging, including optical coherence tomography angiography. The DNA of the first proband was screened using a targeted ocular gene panel, while, in the second proband, direct sequencing of the TIMP3 coding region was performed. Sanger sequencing was also used for segregation analysis within the families. All the previously reported TIMP3 variants were reviewed using the American College of Medical Genetics and the Association for Molecular Pathology interpretation framework. A novel heterozygous variant, c.455A>G p.(Tyr152Cys), in TIMP3 was identified in both families and potentially de novo in one. Optical coherence tomography angiography documented in one patient the development of a choroidal neovascular membrane at 54 years. Including this study, 23 heterozygous variants in TIMP3 have been reported as disease-causing. Application of gene-specific criteria denoted eleven variants as pathogenic, eleven as likely pathogenic, and one as a variant of unknown significance. Our study expands the spectrum of TIMP3 pathogenic variants and highlights the importance of optical coherence tomography angiography for early detection of choroidal neovascular membranes.


Asunto(s)
Neovascularización Coroidal , Degeneración Macular , Humanos , República Checa , Ojo , Mutación , Inhibidor Tisular de Metaloproteinasa-3/genética
4.
Hum Mol Genet ; 30(17): 1591-1606, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34046667

RESUMEN

The forkhead transcription factor FOXE3 is critical for vertebrate eye development. Recessive and dominant variants cause human ocular disease but the full range of phenotypes and mechanisms of action for the two classes of variants are unknown. We identified FOXE3 variants in individuals with congenital eye malformations and carried out in vitro functional analysis on selected alleles. Sixteen new recessive and dominant families, including six novel variants, were identified. Analysis of new and previously reported genetic and clinical data demonstrated a broad phenotypic range with an overlap between recessive and dominant disease. Most families with recessive alleles, composed of truncating and forkhead-domain missense variants, had severe corneal opacity (90%; sclerocornea in 47%), aphakia (83%) and microphthalmia (80%), but some had milder features including isolated cataract. The phenotype was most variable for recessive missense variants, suggesting that the functional consequences may be highly dependent on the type of amino acid substitution and its position. When assessed, aniridia or iris hypoplasia were noted in 89% and optic nerve anomalies in 60% of recessive cases, indicating that these defects are also common and may be underrecognized. In dominant pedigrees, caused by extension variants, normal eye size (96%), cataracts (99%) and variable anterior segment anomalies were seen in most, but some individuals had microphthalmia, aphakia or sclerocornea, more typical of recessive disease. Functional studies identified variable effects on the protein stability, DNA binding, nuclear localization and transcriptional activity for recessive FOXE3 variants, whereas dominant alleles showed severe impairment in all areas and dominant-negative characteristics.


Asunto(s)
Anomalías del Ojo/genética , Ojo/embriología , Factores de Transcripción Forkhead/genética , Adolescente , Alelos , Catarata/genética , Niño , Opacidad de la Córnea/genética , Discapacidades del Desarrollo/genética , Ojo/crecimiento & desarrollo , Anomalías del Ojo/enzimología , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Masculino , Mutación , Linaje , Fenotipo
5.
Clin Genet ; 104(4): 418-426, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37321975

RESUMEN

Four members of a three-generation Czech family with early-onset chorioretinal dystrophy were shown to be heterozygous carriers of the n.37C>T in MIR204. The identification of this previously reported pathogenic variant confirms the existence of a distinct clinical entity caused by a sequence change in MIR204. Chorioretinal dystrophy was variably associated with iris coloboma, congenital glaucoma, and premature cataracts extending the phenotypic range of the condition. In silico analysis of the n.37C>T variant revealed 713 novel targets. Additionally, four family members were shown to be affected by albinism resulting from biallelic pathogenic OCA2 variants. Haplotype analysis excluded relatedness with the original family reported to harbour the n.37C>T variant in MIR204. Identification of a second independent family confirms the existence of a distinct MIR204-associated clinical entity and suggests that the phenotype may also involve congenital glaucoma.


Asunto(s)
Catarata , Coloboma , Glaucoma , MicroARNs , Humanos , Coloboma/complicaciones , Coloboma/genética , Mutación , Linaje , Iris/anomalías , Glaucoma/complicaciones , Glaucoma/genética , Catarata/genética , Catarata/congénito
6.
Am J Hum Genet ; 102(3): 447-459, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29499165

RESUMEN

In a large family of Czech origin, we mapped a locus for an autosomal-dominant corneal endothelial dystrophy, posterior polymorphous corneal dystrophy 4 (PPCD4), to 8q22.3-q24.12. Whole-genome sequencing identified a unique variant (c.20+544G>T) in this locus, within an intronic regulatory region of GRHL2. Targeted sequencing identified the same variant in three additional previously unsolved PPCD-affected families, including a de novo occurrence that suggests this is a recurrent mutation. Two further unique variants were identified in intron 1 of GRHL2 (c.20+257delT and c.20+133delA) in unrelated PPCD-affected families. GRHL2 is a transcription factor that suppresses epithelial-to-mesenchymal transition (EMT) and is a direct transcriptional repressor of ZEB1. ZEB1 mutations leading to haploinsufficiency cause PPCD3. We previously identified promoter mutations in OVOL2, a gene not normally expressed in the corneal endothelium, as the cause of PPCD1. OVOL2 drives mesenchymal-to-epithelial transition (MET) by directly inhibiting EMT-inducing transcription factors, such as ZEB1. Here, we demonstrate that the GRHL2 regulatory variants identified in PPCD4-affected individuals induce increased transcriptional activity in vitro. Furthermore, although GRHL2 is not expressed in corneal endothelial cells in control tissue, we detected GRHL2 in the corneal "endothelium" in PPCD4 tissue. These cells were also positive for epithelial markers E-Cadherin and Cytokeratin 7, indicating they have transitioned to an epithelial-like cell type. We suggest that mutations inducing MET within the corneal endothelium are a convergent pathogenic mechanism leading to dysfunction of the endothelial barrier and disease.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , Proteínas de Unión al ADN/genética , Mutación/genética , Factores de Transcripción/genética , Secuencia de Bases , ADN Intergénico/genética , Endotelio Corneal/patología , Familia , Femenino , Sitios Genéticos , Células HEK293 , Humanos , Intrones/genética , Masculino , Modelos Genéticos , Linaje , Regiones Promotoras Genéticas/genética , Transcripción Genética , Secuenciación Completa del Genoma
7.
Am J Hum Genet ; 102(4): 528-539, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29526280

RESUMEN

Fuchs endothelial corneal dystrophy (FECD) is a common disease for which corneal transplantation is the only treatment option in advanced stages, and alternative treatment strategies are urgently required. Expansion (≥50 copies) of a non-coding trinucleotide repeat in TCF4 confers >76-fold risk for FECD in our large cohort of affected individuals. An FECD subject-derived corneal endothelial cell (CEC) model was developed to probe disease mechanism and investigate therapeutic approaches. The CEC model demonstrated that the repeat expansion leads to nuclear RNA foci, with the sequestration of splicing factor proteins (MBNL1 and MBNL2) to the foci and altered mRNA processing. Antisense oligonucleotide (ASO) treatment led to a significant reduction in the incidence of nuclear foci, MBNL1 recruitment to the foci, and downstream aberrant splicing events, suggesting functional rescue. This proof-of-concept study highlights the potential of a targeted ASO therapy to treat the accessible and tractable corneal tissue affected by this repeat expansion-mediated disease.


Asunto(s)
Distrofia Endotelial de Fuchs/genética , Predisposición Genética a la Enfermedad , Oligonucleótidos Antisentido/farmacología , Factor de Transcripción 4/genética , Expansión de Repetición de Trinucleótido/genética , Anciano , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Estudios de Cohortes , Células Endoteliales/metabolismo , Endotelio Corneal/patología , Femenino , Distrofia Endotelial de Fuchs/patología , Humanos , Masculino , Ratones Endogámicos C57BL , Especificidad de Órganos , Precursores del ARN/genética , Procesamiento Postranscripcional del ARN , Factores de Empalme de ARN/metabolismo , ARN Mensajero/metabolismo , Factores de Riesgo
8.
Genet Med ; 22(7): 1235-1246, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32307445

RESUMEN

PURPOSE: Missing heritability in human diseases represents a major challenge, and this is particularly true for ABCA4-associated Stargardt disease (STGD1). We aimed to elucidate the genomic and transcriptomic variation in 1054 unsolved STGD and STGD-like probands. METHODS: Sequencing of the complete 128-kb ABCA4 gene was performed using single-molecule molecular inversion probes (smMIPs), based on a semiautomated and cost-effective method. Structural variants (SVs) were identified using relative read coverage analyses and putative splice defects were studied using in vitro assays. RESULTS: In 448 biallelic probands 14 known and 13 novel deep-intronic variants were found, resulting in pseudoexon (PE) insertions or exon elongations in 105 alleles. Intriguingly, intron 13 variants c.1938-621G>A and c.1938-514G>A resulted in dual PE insertions consisting of the same upstream, but different downstream PEs. The intron 44 variant c.6148-84A>T resulted in two PE insertions and flanking exon deletions. Eleven distinct large deletions were found, two of which contained small inverted segments. Uniparental isodisomy of chromosome 1 was identified in one proband. CONCLUSION: Deep sequencing of ABCA4 and midigene-based splice assays allowed the identification of SVs and causal deep-intronic variants in 25% of biallelic STGD1 cases, which represents a model study that can be applied to other inherited diseases.


Asunto(s)
Degeneración Macular , Transcriptoma , Transportadoras de Casetes de Unión a ATP/genética , Genómica , Humanos , Intrones , Degeneración Macular/genética , Mutación , Linaje , Enfermedad de Stargardt
9.
Am J Hum Genet ; 98(1): 75-89, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26749309

RESUMEN

Congenital hereditary endothelial dystrophy 1 (CHED1) and posterior polymorphous corneal dystrophy 1 (PPCD1) are autosomal-dominant corneal endothelial dystrophies that have been genetically mapped to overlapping loci on the short arm of chromosome 20. We combined genetic and genomic approaches to identify the cause of disease in extensive pedigrees comprising over 100 affected individuals. After exclusion of pathogenic coding, splice-site, and copy-number variations, a parallel approach using targeted and whole-genome sequencing facilitated the identification of pathogenic variants in a conserved region of the OVOL2 proximal promoter sequence in the index families (c.-339_361dup for CHED1 and c.-370T>C for PPCD1). Direct sequencing of the OVOL2 promoter in other unrelated affected individuals identified two additional mutations within the conserved proximal promoter sequence (c.-274T>G and c.-307T>C). OVOL2 encodes ovo-like zinc finger 2, a C2H2 zinc-finger transcription factor that regulates mesenchymal-to-epithelial transition and acts as a direct transcriptional repressor of the established PPCD-associated gene ZEB1. Interestingly, we did not detect OVOL2 expression in the normal corneal endothelium. Our in vitro data demonstrate that all four mutated OVOL2 promoters exhibited more transcriptional activity than the corresponding wild-type promoter, and we postulate that the mutations identified create cryptic cis-acting regulatory sequence binding sites that drive aberrant OVOL2 expression during endothelial cell development. Our data establish CHED1 and PPCD1 as allelic conditions and show that CHED1 represents the extreme of what can be considered a disease spectrum. They also implicate transcriptional dysregulation of OVOL2 as a common cause of dominantly inherited corneal endothelial dystrophies.


Asunto(s)
Alelos , Distrofias Hereditarias de la Córnea/genética , Mutación , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Secuencia de Bases , ADN , Femenino , Humanos , Masculino , Linaje , Homología de Secuencia de Ácido Nucleico
10.
Exp Eye Res ; 182: 160-166, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30851240

RESUMEN

The aim of this study was to identify the molecular genetic cause of disease in posterior polymorphous corneal dystrophy (PPCD) probands of diverse origin and to assess the utility of massively parallel sequencing in the detection of ZEB1 mutations. We investigated a total of 12 families (five British, four Czech, one Slovak and two Swiss). Ten novel and two recurrent disease-causing mutations in ZEB1, were identified in probands by Sanger (n = 5), exome (n = 4) and genome (n = 3) sequencing. Sanger sequencing was used to confirm the mutations detected by massively parallel sequencing, and to perform segregation analysis. Genome sequencing revealed that one proband harboured a novel ∼0.34 Mb heterozygous de novo deletion spanning exons 1-7 and part of exon 8. Transcript analysis confirmed that the ZEB1 transcript is detectable in blood-derived RNA samples and that the disease-associated variant c.482-2A>G leads to aberrant pre-mRNA splicing. De novo mutations, which are a feature of PPCD3, were found in the current study with an incidence rate of at least 16.6%. In general, massively parallel sequencing is a time-efficient way to detect PPCD3-associated mutations and, importantly, genome sequencing enables the identification of full or partial heterozygous ZEB1 deletions that can evade detection by both Sanger and exome sequencing. These findings contribute to our understanding of PPCD3, for which currently, 49 pathogenic variants have been identified, all of which are predicted to be null alleles.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , ADN/genética , Mutación , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Adolescente , Adulto , Anciano , Secuencia de Bases , Niño , Preescolar , Distrofias Hereditarias de la Córnea/diagnóstico , Distrofias Hereditarias de la Córnea/metabolismo , Análisis Mutacional de ADN , Exones , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Persona de Mediana Edad , Linaje , Eliminación de Secuencia , Adulto Joven , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Dedos de Zinc
11.
BMC Ophthalmol ; 18(1): 250, 2018 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-30223810

RESUMEN

BACKGROUND: The purpose of this study was to identify the genetic cause and describe the clinical phenotype of Schnyder corneal dystrophy (SCD) in six unrelated probands. METHODS: We identified two white Czech, two white British and two South Asian families with a clinical diagnosis of SCD. Ophthalmic assessment included spectral domain optical coherence tomography (SD-OCT) of one individual with advanced disease, and SD-OCT and confocal microscopy of a child with early stages of disease. UBIAD1 coding exons were amplified and Sanger sequenced in each proband. A fasting serum lipid profile was measured in three probands. Paternity testing was performed in one family. RESULTS: A novel heterozygous c.527G>A; p.(Gly176Glu) mutation in UBIAD1 was identified in one Czech proband. In the second Czech proband, aged 6 years when first examined, a previously described de novo heterozygous c.289G>A; p.(Ala97Thr) mutation was found. Two probands of South Asian descent carried a known c.305G>A; p.(Asn102Ser) mutation in the heterozygous state. Previously reported heterozygous c.361C>T; p.(Leu121Phe) and c.308C>T; p.(Thr103Ile) mutations were found in two white British families. Although crystalline deposits were present in all probands the affected area was small in some individuals. Corneal arcus and stromal haze were the most prominent phenotypical feature in two probands. In the Czech probands, SD-OCT confirmed accumulation of reflective material in the anterior stroma. Crystalline deposits were visualized by confocal microscopy. Mild dyslipidemia was found in all three individuals tested. CONCLUSION: Although de novo occurrence of mutations in UBIAD1 is extremely rare, SCD should be considered in the differential diagnosis of bilateral corneal haze and/or crystal deposition, especially in children.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , ADN/genética , Dimetilaliltranstransferasa/genética , Mutación , Adulto , Niño , Distrofias Hereditarias de la Córnea/diagnóstico , Distrofias Hereditarias de la Córnea/metabolismo , Análisis Mutacional de ADN , Dimetilaliltranstransferasa/metabolismo , Femenino , Humanos , Masculino , Microscopía Confocal , Persona de Mediana Edad , Linaje , Fenotipo , Tomografía de Coherencia Óptica/métodos
12.
Mol Biol Rep ; 44(6): 435-440, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28849415

RESUMEN

A bilaterally blind woman, with a three generation family history of autosomal dominant congenital cataracts, variably associated with iris colobomata and microcornea, sought preconception genetic consultation. Whole-exome sequencing was performed in three affected family members, one unaffected first degree relative, and one spouse. The sequence variant c.168C>G; p.(Tyr56∗) in CRYGD, previously reported as pathogenic, and a novel mutation c.809C>A; p.(Ser270Tyr) in MAF, were identified in two affected family members; the grandmother, and half-brother of the proband. The proband inherited only the MAF mutation, whereas her clinically unaffected sister had the CRYGD change. In silico analysis supported a pathogenic role of p.(Ser270Tyr) in MAF, which was absent from publicly available whole-exome datasets, and 1161 Czech individuals. The frequency of CRYGD p.(Tyr56∗) in the ExAC dataset was higher than the estimated incidence of congenital cataract in the general population. Our study highlights that patients with genetically heterogeneous conditions may exhibit rare variants in more than one disease-associated gene, warranting caution with data interpretation, and supporting parallel screening of all genes known to harbour pathogenic mutations for a given phenotype. The pathogenicity of sequence variants previously reported as cataract-causing may require re-assessment in light of recently released datasets of human genomic variation.


Asunto(s)
Catarata/genética , Proteínas Proto-Oncogénicas c-maf/genética , gamma-Cristalinas/genética , Adulto , Catarata/congénito , Análisis Mutacional de ADN/métodos , Exoma/genética , Femenino , Genes Dominantes/genética , Humanos , Masculino , Mutación/genética , Linaje , Fenotipo , Proteínas Proto-Oncogénicas c-maf/metabolismo , Secuenciación del Exoma/métodos , gamma-Cristalinas/metabolismo
13.
Mol Biol Rep ; 44(6): 441, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29019065

RESUMEN

There was a spacing error in the initial online publication, and there were errors in the Acknowledgments section. The original article has been updated.

14.
Am J Med Genet A ; 170(7): 1843-8, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27256633

RESUMEN

We observed bilateral cataracts on second trimester ultrasound, in two consecutive pregnancies, with no other structural defects detected. The parents were unrelated and had no family history for the disease. The first pregnancy was terminated in week 22. Copy number variation analysis revealed, in both the aborted fetus and the mother, a 495 kb duplication at 22q11.23 encompassing CRYBB3 and CRYBB2, and not present in variation databases. In the second pregnancy, lens hyperechogenicity was detected by ultrasound at week 13 and 4 days. The identical duplication at 22q11.23 was found in the fetus and considered as possibly pathogenic. At weeks 22 and 30, smaller orbit measurements were elucidated on ultrasound, raising concerns as to the underlying molecular genetic cause, necessitating further investigation. Whole-exome sequencing, using DNA of the first fetus, was performed shortly after the birth of a male child, and two truncating RAB3GAP1 mutations were detected: c.538G>T; p. (Glu180*) and c.943C>T; p. (Arg315*). Neither mutation has been previously reported to be disease-causing; however, evaluation in the context of previously published literature indicated their deleterious nature, implying a clinical diagnosis of Warburg micro syndrome or Martsolf syndrome. Sanger sequencing confirmed segregation of the two mutations within the family, consistent with autosomal recessive inheritance. The child born from the second pregnancy showed features typical of Warburg micro syndrome, with the exception of microcephaly, at age 31 months. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Anomalías Múltiples/genética , Catarata/congénito , Catarata/genética , Córnea/anomalías , Hipogonadismo/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Atrofia Óptica/genética , Cadena B de beta-Cristalina/genética , Proteínas de Unión al GTP rab3/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/fisiopatología , Feto Abortado/fisiopatología , Catarata/diagnóstico , Catarata/fisiopatología , Córnea/fisiopatología , Variaciones en el Número de Copia de ADN/genética , Exones/genética , Femenino , Humanos , Hipogonadismo/diagnóstico , Hipogonadismo/fisiopatología , Lactante , Recién Nacido , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/fisiopatología , Masculino , Microcefalia/diagnóstico , Microcefalia/fisiopatología , Mutación , Atrofia Óptica/diagnóstico , Atrofia Óptica/fisiopatología , Linaje , Embarazo , Análisis de Secuencia de ADN , Ultrasonografía Prenatal
15.
Graefes Arch Clin Exp Ophthalmol ; 254(9): 1833-9, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27113771

RESUMEN

PURPOSE: The aim of this study was to determine the molecular genetic basis of an early-onset severe retinal dystrophy in three unrelated consecutive patients of Czech origin and to describe their ocular phenotype. METHODS: DNA samples from two probands were analyzed using a genotyping microarray (Asper) followed by either target analysis of 43 genes implicated in retinal disorders by next generation sequencing or whole-exome sequencing, respectively. The third proband underwent conventional Sanger sequencing of CRB1 based on her ocular findings. RESULTS: All three probands harboured a known disease-causing mutation c.2843G>A; p.(Cys948Tyr) in the CRB1 gene. One individual was homozygous for this mutation, while in the other two probands c.2308G>A; p.(Gly770Ser) and c.3121A>G; p.(Met1041Val) were also identified in the heterozygous state, respectively. Both variants were novel and evaluated by in silico analysis as pathogenic. A false-negative result was observed in one of the two samples examined by the genotyping microarray. Disease onset in all patients was before the age of 7 years. Hypermetropic refractive error, bilateral nummular retinal pigmentation, retinal thickening and cystoid spaces in the macula were observed in two probands, aged 6 and 7 years. The third proband, aged 28 years, had bone spicule-like pigmentary changes associated with increased retinal nerve fiber layer. CONCLUSIONS: The first study reporting on the molecular genetic cause of non-syndromic early-onset severe retinal dystrophy in Czech patients identified one homozygous and two compound heterozygote probands with CRB1 mutations. Retina nerve fibre layer measurements should be considered an integral part of the clinical evaluation of retinal dystrophies. Detailed clinical examination and imaging can both direct molecular screening and help to confirm or refute disease causation of identified variants.


Asunto(s)
Enfermedades Hereditarias del Ojo/genética , Proteínas del Ojo/genética , Proteínas de la Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Retina/diagnóstico por imagen , Distrofias Retinianas/genética , Adolescente , Adulto , Niño , Análisis Mutacional de ADN , Electrorretinografía , Enfermedades Hereditarias del Ojo/diagnóstico , Enfermedades Hereditarias del Ojo/metabolismo , Proteínas del Ojo/metabolismo , Femenino , Genotipo , Homocigoto , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Linaje , Fenotipo , Retina/metabolismo , Retina/fisiopatología , Distrofias Retinianas/diagnóstico , Distrofias Retinianas/metabolismo , Adulto Joven
16.
Ann Hum Genet ; 79(1): 1-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25441224

RESUMEN

Posterior polymorphous corneal dystrophy 3 (PPCD3) is a rare autosomal dominant disorder caused by mutations in ZEB1. To date all identified disease-causing variants were unique to the studied families, except for c.1576dup. We have detected six novel ZEB1 mutations; c.1749_1750del; p.(Pro584*) and c.1717_1718del; p.(Val573Phefs*12) in two Czech families, c.1176dup; p.(Ala393Serfs*19), c.1100C>A; p.(Ser367*), c.627del; p.(Phe209Leufs*11) in three British families and a splice site mutation, c.685-2A>G, in a patient of Sri Lankan origin. An additional British proband had the c.1576dup; p.(Val526Glyfs*3) mutation previously reported in other populations. Clinical findings were variable and included bilateral congenital corneal opacity in one proband, development of opacity before the age of 2 years in another individual and bilateral iris flocculi in yet another subject. The majority of eyes examined by corneal topography (10 out of 16) had an abnormally steep cornea (flat keratometry 46.5-52.7 diopters, steep keratometry 48.1-54.0 diopters). One proband underwent surgery for cryptorchidism. Our study further demonstrates that PPCD3 can present as corneal edema in early childhood, and that an abnormally steep keratometry is a common feature of this condition. As cryptorchidism has been previously observed in two other PPCD3 cases, its association with the disease warrants further investigation.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Adolescente , Adulto , Niño , Codón sin Sentido , Análisis Mutacional de ADN , Femenino , Mutación del Sistema de Lectura , Humanos , Masculino , Linaje , Fenotipo , Sitios de Empalme de ARN/genética , Eliminación de Secuencia , Adulto Joven , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
17.
Ophthalmic Res ; 53(1): 30-5, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25500497

RESUMEN

BACKGROUND/AIMS: To identify the underlying molecular genetic cause of disease in a patient with Harboyan syndrome and to perform a detailed assessment of her renal function. We also assessed the influence of the SLC4A11 mutation identified on the corneal endothelium in the heterozygous state. METHODS: A 55-year-old female was examined ophthalmologically, audiologically and nephrologically including 24-hour urine collection. The coding region of SLC4A11 was directly sequenced. Specular microscopy was performed in the proband's 21-year-old daughter. RESULTS: The proband had bilateral iridectomy at the age of 3 months because of an initial diagnosis of congenital glaucoma and since the age of 12 years she underwent several keratoplasties in each eye. Nephrological examination did not reveal any abnormalities. Moderate bilateral sensorineural hearing loss was confirmed by audiometry. A novel homozygous mutation predicted to lead to a premature stop codon at the protein level, c.2188C>T; p.(Arg730*), was identified in SLC4A11. No changes in corneal endothelial cell morphology or density were observed in the heterozygous daughter. CONCLUSION: In contrast to the Slc4a11(-/-) mouse, no abnormalities in daily renal ion excretion or polyuria were observed in the Harboyan syndrome patient. The mutation identified does not affect corneal endothelial cell morphology or density in the heterozygous state.


Asunto(s)
Proteínas de Transporte de Anión/genética , Antiportadores/genética , Codón sin Sentido , Distrofias Hereditarias de la Córnea/genética , Pérdida Auditiva Sensorineural/genética , Riñón/fisiología , Audiometría , Distrofias Hereditarias de la Córnea/diagnóstico , Distrofias Hereditarias de la Córnea/fisiopatología , Paquimetría Corneal , Análisis Mutacional de ADN , Endotelio Corneal/patología , Femenino , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Pruebas de Función Renal , Pruebas de Función Hepática , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Agudeza Visual/fisiología , Adulto Joven
18.
Genes (Basel) ; 15(6)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38927735

RESUMEN

The aim of this study was to describe the clinical and molecular genetic findings in seven individuals from three unrelated families with Blau syndrome. A complex ophthalmic and general health examination including diagnostic imaging was performed. The NOD2 mutational hot spot located in exon 4 was Sanger sequenced in all three probands. Two individuals also underwent autoinflammatory disorder gene panel screening, and in one subject, exome sequencing was performed. Blau syndrome presenting as uveitis, skin rush or arthritis was diagnosed in four cases from three families. In two individuals from one family, only camptodactyly was noted, while another member had camptodactyly in combination with non-active uveitis and angioid streaks. One proband developed two attacks of meningoencephalitis attributed to presumed neurosarcoidosis, which is a rare finding in Blau syndrome. The probands from families 1 and 2 carried pathogenic variants in NOD2 (NM_022162.3): c.1001G>A p.(Arg334Gln) and c.1000C>T p.(Arg334Trp), respectively. In family 3, two variants of unknown significance in a heterozygous state were found: c.1412G>T p.(Arg471Leu) in NOD2 and c.928C>T p.(Arg310*) in NLRC4 (NM_001199139.1). In conclusion, Blau syndrome is a phenotypically highly variable, and there is a need to raise awareness about all clinical manifestations, including neurosarcoidosis. Variants of unknown significance pose a significant challenge regarding their contribution to etiopathogenesis of autoinflammatory diseases.


Asunto(s)
Artritis , Mutación , Proteína Adaptadora de Señalización NOD2 , Linaje , Sarcoidosis , Sinovitis , Uveítis , Humanos , Artritis/genética , Artritis/diagnóstico , Artropatía Neurógena/genética , Artropatía Neurógena/diagnóstico , Enfermedades del Sistema Nervioso Central , Secuenciación del Exoma , Enfermedades Autoinflamatorias Hereditarias , Linfedema/genética , Linfedema/diagnóstico , Proteína Adaptadora de Señalización NOD2/genética , Sarcoidosis/genética , Sarcoidosis/diagnóstico , Sinovitis/genética , Sinovitis/diagnóstico , Uveítis/genética , Uveítis/diagnóstico
19.
Biomolecules ; 14(3)2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38540785

RESUMEN

Inherited macular dystrophies (iMDs) are a group of genetic disorders, which affect the central region of the retina. To investigate the genetic basis of iMDs, we used single-molecule Molecular Inversion Probes to sequence 105 maculopathy-associated genes in 1352 patients diagnosed with iMDs. Within this cohort, 39.8% of patients were considered genetically explained by 460 different variants in 49 distinct genes of which 73 were novel variants, with some affecting splicing. The top five most frequent causative genes were ABCA4 (37.2%), PRPH2 (6.7%), CDHR1 (6.1%), PROM1 (4.3%) and RP1L1 (3.1%). Interestingly, variants with incomplete penetrance were revealed in almost one-third of patients considered solved (28.1%), and therefore, a proportion of patients may not be explained solely by the variants reported. This includes eight previously reported variants with incomplete penetrance in addition to CDHR1:c.783G>A and CNGB3:c.1208G>A. Notably, segregation analysis was not routinely performed for variant phasing-a limitation, which may also impact the overall diagnostic yield. The relatively high proportion of probands without any putative causal variant (60.2%) highlights the need to explore variants with incomplete penetrance, the potential modifiers of disease and the genetic overlap between iMDs and age-related macular degeneration. Our results provide valuable insights into the genetic landscape of iMDs and warrant future exploration to determine the involvement of other maculopathy genes.


Asunto(s)
Degeneración Macular , Humanos , Mutación , Penetrancia , Linaje , Degeneración Macular/genética , Retina , Fenotipo , Transportadoras de Casetes de Unión a ATP/genética , Proteínas del Ojo , Proteínas Relacionadas con las Cadherinas , Proteínas del Tejido Nervioso/genética
20.
J Neural Transm (Vienna) ; 120(6): 977-82, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23653221

RESUMEN

Keratoconus (KC) is an eye disease characterized by the progressive thinning and protrusion of the cornea, which results in the loss of visual acuity. This disorder remains poorly understood, although recent studies indicate the involvement of genetic and environmental factors. Recently, we have found that the distribution of the cross-linking enzyme lysyl oxidase (LOX) is markedly decreased in about 63 % of keratoconic specimens. Similarly, LOX activity is significantly reduced by 38 % compared to control tissue. Nearly 70 systemic disorders have been reported in association with KC, most of them affecting the extracellular matrix. In this review we attempted to ascertain whether any KC-associated diseases exhibit signs that may reflect LOX impairment. We hypothesized that very similar changes in the extracellular matrix, particularly at the level of collagen metabolism, including LOX impairment in mitral leaflets, may reflect an association between KC and mitral valve prolapse. Moreover, this putative association is supported by the high frequency of Down syndrome in both diseases. Among other disorders that have been found to coincide with KC, we did not find any in which the LOX enzyme may be directly or indirectly impaired. On the other hand, in cases where KC is present along with other connective tissue disorders (Marfan syndrome, Ehlers-Danlos syndrome and others), KC may not arise as a localized manifestation, but rather may be induced as the result of a more complex connective tissue disorder.


Asunto(s)
Queratocono/enzimología , Proteína-Lisina 6-Oxidasa/metabolismo , Enfermedades del Tejido Conjuntivo/complicaciones , Humanos , Queratocono/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA