Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Bases 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.
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
3.
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
4.
Am J Hum Genet ; 99(6): 1338-1352, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27839872

RESUMEN

Anterior segment dysgeneses (ASDs) comprise a spectrum of developmental disorders affecting the anterior segment of the eye. Here, we describe three unrelated families affected by a previously unclassified form of ASD. Shared ocular manifestations include bilateral iris hypoplasia, ectopia lentis, corectopia, ectropion uveae, and cataracts. Whole-exome sequencing and targeted Sanger sequencing identified mutations in CPAMD8 (C3 and PZP-like alpha-2-macroglobulin domain-containing protein 8) as the cause of recessive ASD in all three families. A homozygous missense mutation in the evolutionarily conserved alpha-2-macroglobulin (A2M) domain of CPAMD8, c.4351T>C (p. Ser1451Pro), was identified in family 1. In family 2, compound heterozygous frameshift, c.2352_2353insC (p.Arg785Glnfs∗23), and splice-site, c.4549-1G>A, mutations were identified. Two affected siblings in the third family were compound heterozygous for splice-site mutations c.700+1G>T and c.4002+1G>A. CPAMD8 splice-site mutations caused aberrant pre-mRNA splicing in vivo or in vitro. Intriguingly, our phylogenetic analysis revealed rodent lineage-specific CPAMD8 deletion, precluding a developmental expression study in mice. We therefore investigated the spatiotemporal expression of CPAMD8 in the developing human eye. RT-PCR and in situ hybridization revealed CPAMD8 expression in the lens, iris, cornea, and retina early in development, including strong expression in the distal tips of the retinal neuroepithelium that form the iris and ciliary body, thus correlating CPAMD8 expression with the affected tissues. Our study delineates a unique form of recessive ASD and defines a role for CPAMD8, a protein of unknown function, in anterior segment development, implying another pathway for the pathogenicity of ASD.


Asunto(s)
Segmento Anterior del Ojo/anomalías , Complemento C3/genética , Anomalías del Ojo/genética , Genes Recesivos/genética , Mutación , Inhibidor de Tripsina Pancreática de Kazal/genética , alfa-Macroglobulinas/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Segmento Anterior del Ojo/metabolismo , Niño , Preescolar , Complemento C3/química , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inhibidor de Tripsina Pancreática de Kazal/química , Adulto Joven , alfa-Macroglobulinas/química
5.
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
6.
Genet Med ; 21(9): 2092-2102, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30733599

RESUMEN

PURPOSE: To demonstrate the utility of an amplification-free long-read sequencing method to characterize the Fuchs endothelial corneal dystrophy (FECD)-associated intronic TCF4 triplet repeat (CTG18.1). METHODS: We applied an amplification-free method, utilizing the CRISPR/Cas9 system, in combination with PacBio single-molecule real-time (SMRT) long-read sequencing, to study CTG18.1. FECD patient samples displaying a diverse range of CTG18.1 allele lengths and zygosity status (n = 11) were analyzed. A robust data analysis pipeline was developed to effectively filter, align, and interrogate CTG18.1-specific reads. All results were compared with conventional polymerase chain reaction (PCR)-based fragment analysis. RESULTS: CRISPR-guided SMRT sequencing of CTG18.1 provided accurate genotyping information for all samples and phasing was possible for 18/22 alleles sequenced. Repeat length instability was observed for all expanded (≥50 repeats) phased CTG18.1 alleles analyzed. Furthermore, higher levels of repeat instability were associated with increased CTG18.1 allele length (mode length ≥91 repeats) indicating that expanded alleles behave dynamically. CONCLUSION: CRISPR-guided SMRT sequencing of CTG18.1 has revealed novel insights into CTG18.1 length instability. Furthermore, this study provides a framework to improve the molecular diagnostic accuracy for CTG18.1-mediated FECD, which we anticipate will become increasingly important as gene-directed therapies are developed for this common age-related and sight threatening disease.


Asunto(s)
Distrofia Endotelial de Fuchs/genética , Predisposición Genética a la Enfermedad , Factor de Transcripción 4/genética , Expansión de Repetición de Trinucleótido/genética , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Sistemas CRISPR-Cas/genética , Femenino , Distrofia Endotelial de Fuchs/patología , Genotipo , Humanos , Intrones/genética , Masculino , Persona de Mediana Edad , Análisis de Secuencia de ADN , Imagen Individual de Molécula , Repeticiones de Trinucleótidos/genética
7.
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
8.
Am J Hum Genet ; 93(2): 321-9, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23849777

RESUMEN

Retinitis pigmentosa (RP) is a genetically heterogeneous retinal degeneration characterized by photoreceptor death, which results in visual failure. Here, we used a combination of homozygosity mapping and exome sequencing to identify mutations in ARL2BP, which encodes an effector protein of the small GTPases ARL2 and ARL3, as causative for autosomal-recessive RP (RP66). In a family affected by RP and situs inversus, a homozygous, splice-acceptor mutation, c.101-1G>C, which alters pre-mRNA splicing of ARLBP2 in blood RNA, was identified. In another family, a homozygous c.134T>G (p.Met45Arg) mutation was identified. In the mouse retina, ARL2BP localized to the basal body and cilium-associated centriole of photoreceptors and the periciliary extension of the inner segment. Depletion of ARL2BP caused cilia shortening. Moreover, depletion of ARL2, but not ARL3, caused displacement of ARL2BP from the basal body, suggesting that ARL2 is vital for recruiting or anchoring ARL2BP at the base of the cilium. This hypothesis is supported by the finding that the p.Met45Arg amino acid substitution reduced binding to ARL2 and caused the loss of ARL2BP localization at the basal body in ciliated nasal epithelial cells. These data demonstrate a role for ARL2BP and ARL2 in primary cilia function and that this role is essential for normal photoreceptor maintenance and function.


Asunto(s)
Factores de Ribosilacion-ADP/genética , Proteínas Portadoras/genética , Proteínas de Unión al GTP/genética , Mutación , Células Fotorreceptoras/metabolismo , Retinitis Pigmentosa/genética , Factores de Ribosilacion-ADP/metabolismo , Adulto , Animales , Secuencia de Bases , Proteínas Portadoras/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Proteínas de Unión al GTP/metabolismo , Genes Recesivos , Homocigoto , Humanos , Masculino , Proteínas de Transporte de Membrana , Ratones , Datos de Secuencia Molecular , Linaje , Células Fotorreceptoras/patología , Unión Proteica , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Factores de Transcripción
9.
Hum Mutat ; 36(4): 463-73, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25676728

RESUMEN

Corneal dystrophies are a clinically and genetically heterogeneous group of inherited disorders that bilaterally affect corneal transparency. They are defined according to the corneal layer affected and by their genetic cause. In this study, we identified a dominantly inherited epithelial recurrent erosion dystrophy (ERED)-like disease that is common in northern Sweden. Whole-exome sequencing resulted in the identification of a novel mutation, c.2816C>T, p.T939I, in the COL17A1 gene, which encodes collagen type XVII alpha 1. The variant segregated with disease in a genealogically expanded pedigree dating back 200 years. We also investigated a unique COL17A1 synonymous variant, c.3156C>T, identified in a previously reported unrelated dominant ERED-like family linked to a locus on chromosome 10q23-q24 encompassing COL17A1. We show that this variant introduces a cryptic donor site resulting in aberrant pre-mRNA splicing and is highly likely to be pathogenic. Bi-allelic COL17A1 mutations have previously been associated with a recessive skin disorder, junctional epidermolysis bullosa, with recurrent corneal erosions being reported in some cases. Our findings implicate presumed gain-of-function COL17A1 mutations causing dominantly inherited ERED and improve understanding of the underlying pathology.


Asunto(s)
Autoantígenos/genética , Distrofias Hereditarias de la Córnea/diagnóstico , Distrofias Hereditarias de la Córnea/genética , Epitelio Corneal/patología , Estudios de Asociación Genética , Mutación , Colágenos no Fibrilares/genética , Adolescente , Adulto , Edad de Inicio , Anciano , Anciano de 80 o más Años , Autoantígenos/metabolismo , Niño , Femenino , Expresión Génica , Variación Genética , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Colágenos no Fibrilares/metabolismo , Linaje , Fenotipo , Polimorfismo de Nucleótido Simple , Empalme del ARN , Adulto Joven , Colágeno Tipo XVII
10.
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
11.
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
12.
Hum Mutat ; 35(3): 289-93, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24375934

RESUMEN

Inherited retinal dystrophies are a major cause of childhood blindness. Here, we describe the identification of a homozygous frameshift mutation (c.1194_1195delAG, p.Arg398Serfs*9) in TUB in a child from a consanguineous UK Caucasian family investigated using autozygosity mapping and whole-exome sequencing. The proband presented with obesity, night blindness, decreased visual acuity, and electrophysiological features of a rod cone dystrophy. The mutation was also found in two of the proband's siblings with retinal dystrophy and resulted in mislocalization of the truncated protein. In contrast to known forms of retinal dystrophy, including those caused by mutations in the tubby-like protein TULP-1, loss of function of TUB in the proband and two affected family members was associated with early-onset obesity, consistent with an additional role for TUB in energy homeostasis.


Asunto(s)
Mutación del Sistema de Lectura , Homocigoto , Obesidad/genética , Proteínas/genética , Retinitis Pigmentosa/genética , Proteínas Adaptadoras Transductoras de Señales , Niño , Mapeo Cromosómico , Consanguinidad , Proteínas del Ojo/genética , Femenino , Genes Recesivos , Homeostasis , Humanos , Masculino , Linaje , Reino Unido , Población Blanca/genética
13.
Hum Mutat ; 35(11): 1354-62, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25168334

RESUMEN

Mutations in the OPN1LW (L-) and OPN1MW (M-)cone opsin genes underlie a spectrum of cone photoreceptor defects from stationary loss of color vision to progressive retinal degeneration. Genotypes of 22 families with a range of cone disorders were grouped into three classes: deletions of the locus control region (LCR); missense mutation (p.Cys203Arg) in an L-/M-hybrid gene; and exon 3 single-nucleotide polymorphism (SNP) interchange haplotypes in an otherwise normal gene array. Moderate-to-high myopia was observed in all mutation categories. Individuals with LCR deletions or p.Cys203Arg mutations were more likely to have nystagmus and poor vision, with disease progression in some p.Cys203Arg patients. Three disease-associated exon 3 SNP haplotypes encoding LIAVA, LVAVA, or MIAVA were identified in our cohort. These patients were less likely to have nystagmus but more likely to show progression, with all patients over the age of 40 years having marked macular abnormalities. Previously, the haplotype LIAVA has been shown to result in exon 3 skipping. Here, we show that haplotypes LVAVA and MIAVA also result in aberrant splicing, with a residual low level of correctly spliced cone opsin. The OPN1LW/OPN1MW:c.532A>G SNP, common to all three disease-associated haplotypes, appears to be principally responsible for this mutational mechanism.


Asunto(s)
Opsinas de los Conos/genética , Estudios de Asociación Genética , Genotipo , Mutación , Fenotipo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Sustitución de Aminoácidos , Niño , Preescolar , Orden Génico , Silenciador del Gen , Enfermedades Genéticas Ligadas al Cromosoma X/diagnóstico , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Haplotipos , Hemicigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Oftalmoscopios , Linaje , Polimorfismo de Nucleótido Simple , Empalme del ARN , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/genética , Eliminación de Secuencia , Adulto Joven
14.
Hum Mol Genet ; 21(16): 3647-54, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22619378

RESUMEN

X-linked retinitis pigmentosa (XLRP) is genetically heterogeneous with two causative genes identified, RPGR and RP2. We previously mapped a locus for a severe form of XLRP, RP23, to a 10.71 Mb interval on Xp22.31-22.13 containing 62 genes. Candidate gene screening failed to identify a causative mutation, so we adopted targeted genomic next-generation sequencing of the disease interval to determine the molecular cause of RP23. No coding variants or variants within or near splice sites were identified. In contrast, a variant deep within intron 9 of OFD1 increased the splice site prediction score 4 bp upstream of the variant. Mutations in OFD1 cause the syndromic ciliopathies orofaciodigital syndrome-1, which is male lethal, Simpson-Golabi-Behmel syndrome type 2 and Joubert syndrome. We tested the effect of the IVS9+706A>G variant on OFD1 splicing in vivo. In RP23 patient-derived RNA, we detected an OFD1 transcript with the insertion of a cryptic exon spliced between exons 9 and 10 causing a frameshift, p.N313fs.X330. Correctly spliced OFD1 was also detected in patient-derived RNA, although at reduced levels (39%), hence the mutation is not male lethal. Our data suggest that photoreceptors are uniquely susceptible to reduced expression of OFD1 and that an alternative disease mechanism can cause XLRP. This disease mechanism of reduced expression for a syndromic ciliopathy gene causing isolated retinal degeneration is reminiscent of CEP290 intronic mutations that cause Leber congenital amaurosis, and we speculate that reduced dosage of correctly spliced ciliopathy genes may be a common disease mechanism in retinal degenerations.


Asunto(s)
Mutación del Sistema de Lectura , Proteínas/genética , Retinitis Pigmentosa/etiología , Secuencia de Aminoácidos , Secuencia de Bases , Cromosomas Humanos X , Exones , Humanos , Intrones , Masculino , Datos de Secuencia Molecular , Sitios de Empalme de ARN , Retinitis Pigmentosa/genética , Análisis de Secuencia de ADN
15.
Am J Hum Genet ; 89(1): 183-90, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21763485

RESUMEN

Inherited retinal degenerations, including retinitis pigmentosa (RP) and Leber congenital amaurosis (LCA), comprise a group of disorders showing high genetic and allelic heterogeneity. The determination of a full catalog of genes that can, when mutated, cause human retinal disease is a powerful means to understand the molecular physiology and pathology of the human retina. As more genes are found, remaining ones are likely to be rarer and/or unexpected candidates. Here, we identify a family in which all known RP/LCA-related genes are unlikely to be associated with their disorder. A combination of homozygosity mapping and exome sequencing identifies a homozygous nonsense mutation, c.496C>T (p.Arg166X), in a gene, KCNJ13, encoding a potassium channel subunit Kir7.1. A screen of a further 333 unrelated individuals with recessive retinal degeneration identified an additional proband, homozygous for a missense mutation, c.722T>C (p.Leu241Pro), in the same gene. The three affected members of the two families have been diagnosed with LCA. All have a distinct and unusual retinal appearance and a similar early onset of visual loss, suggesting both impaired retinal development and progressive retinal degeneration, involving both rod and cone pathways. Examination of heterozygotes revealed no ocular disease. This finding implicates Kir7.1 as having an important role in human retinal development and maintenance. This disorder adds to a small diverse group of diseases consequent upon loss or reduced function of inwardly rectifying potassium channels affecting various organs. The distinct retinal phenotype that results from biallelic mutations in KCNJ13 should facilitate the molecular diagnosis in further families.


Asunto(s)
Genes Recesivos , Amaurosis Congénita de Leber/genética , Canales de Potasio de Rectificación Interna/genética , Adulto , Secuencia de Aminoácidos , Codón sin Sentido , Exones , Femenino , Variación Genética , Homocigoto , Humanos , Amaurosis Congénita de Leber/patología , Masculino , Datos de Secuencia Molecular , Ceguera Nocturna/genética , Linaje , Canales de Potasio/genética , Canales de Potasio de Rectificación Interna/metabolismo , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología
16.
Am J Hum Genet ; 89(6): 782-91, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22137173

RESUMEN

Flecked-retina syndromes, including fundus flavimaculatus, fundus albipunctatus, and benign fleck retina, comprise a group of disorders with widespread or limited distribution of yellow-white retinal lesions of various sizes and configurations. Three siblings who have benign fleck retina and were born to consanguineous parents are the basis of this report. A combination of homozygosity mapping and exome sequencing helped to identify a homozygous missense mutation, c.133G>T (p.Gly45Cys), in PLA2G5, a gene encoding a secreted phospholipase (group V phospholipase A(2)). A screen of a further four unrelated individuals with benign fleck retina detected biallelic variants in the same gene in three patients. In contrast, no loss of function or common (minor-allele frequency>0.05%) nonsynonymous PLA2G5 variants have been previously reported (EVS, dbSNP, 1000 Genomes Project) or were detected in an internal database of 224 exomes (from subjects with adult onset neurodegenerative disease and without a diagnosis of ophthalmic disease). All seven affected individuals had fundoscopic features compatible with those previously described in benign fleck retina and no visual or electrophysiological deficits. No medical history of major illness was reported. Levels of low-density lipoprotein were mildly elevated in two patients. Optical coherence tomography and fundus autofluorescence findings suggest that group V phospholipase A(2) plays a role in the phagocytosis of photoreceptor outer-segment discs by the retinal pigment epithelium. Surprisingly, immunohistochemical staining of human retinal tissue revealed localization of the protein predominantly in the inner and outer plexiform layers.


Asunto(s)
Anomalías del Ojo/genética , Fosfolipasas A2 Grupo V/genética , Homocigoto , Mutación Missense , Retina/anomalías , Adulto , Anciano de 80 o más Años , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , Niño , Consanguinidad , Femenino , Estudios de Asociación Genética , Fosfolipasas A2 Grupo V/metabolismo , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Polimorfismo de Nucleótido Simple , Transporte de Proteínas , Retina/metabolismo
17.
Ophthalmology ; 121(1): 234-245, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24148654

RESUMEN

PURPOSE: To characterize retinal structure and function in achromatopsia (ACHM) in preparation for clinical trials of gene therapy. DESIGN: Cross-sectional study. PARTICIPANTS: Forty subjects with ACHM. METHODS: All subjects underwent spectral domain optical coherence tomography (SD-OCT), microperimetry, and molecular genetic testing. Foveal structure on SD-OCT was graded into 5 distinct categories: (1) continuous inner segment ellipsoid (ISe), (2) ISe disruption, (3) ISe absence, (4) presence of a hyporeflective zone (HRZ), and (5) outer retinal atrophy including retinal pigment epithelial loss. Foveal and outer nuclear layer (ONL) thickness was measured and presence of hypoplasia determined. MAIN OUTCOME MEASURES: Photoreceptor appearance on SD-OCT imaging, foveal and ONL thickness, presence of foveal hypoplasia, retinal sensitivity and fixation stability, and association of these parameters with age and genotype. RESULTS: Forty subjects with a mean age of 24.9 years (range, 6-52 years) were included. Disease-causing variants were found in CNGA3 (n = 18), CNGB3 (n = 15), GNAT2 (n = 4), and PDE6C (n = 1). No variants were found in 2 individuals. In all, 22.5% of subjects had a continuous ISe layer at the fovea, 27.5% had ISe disruption, 20% had an absent ISe layer, 22.5% had an HRZ, and 7.5% had outer retinal atrophy. No significant differences in age (P = 0.77), mean retinal sensitivity (P = 0.21), or fixation stability (P = 0.34) across the 5 SD-OCT categories were evident. No correlation was found between age and foveal thickness (P = 0.84) or between age and foveal ONL thickness (P = 0.12). CONCLUSIONS: The lack of a clear association of disruption of retinal structure or function in ACHM with age suggests that the window of opportunity for intervention by gene therapy is wider in some individuals than previously indicated. Therefore, the potential benefit for a given subject is likely to be better predicted by specific measurement of photoreceptor structure rather than simply by age. The ability to directly assess cone photoreceptor preservation with SD-OCT and/or adaptive optics imaging is likely to prove invaluable in selecting subjects for future trials and measuring the trials' impact.


Asunto(s)
Defectos de la Visión Cromática/fisiopatología , Retina/fisiopatología , Adolescente , Adulto , Niño , Defectos de la Visión Cromática/diagnóstico , Defectos de la Visión Cromática/genética , Estudios Transversales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Proteínas del Ojo/genética , Femenino , Estudios de Asociación Genética , Terapia Genética , Proteínas de Unión al GTP Heterotriméricas/genética , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología , Pruebas del Campo Visual , Campos Visuales/fisiología , Adulto Joven
18.
Eur J Hum Genet ; 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39169229

RESUMEN

Corneal dystrophies are phenotypically and genetically heterogeneous, often resulting in visual impairment caused by corneal opacification. We investigated the genetic cause of an autosomal dominant corneal stromal dystrophy in a pedigree with eight affected individuals in three generations. Affected individuals had diffuse central stromal opacity, with reduced visual acuity in older family members. Histopathology of affected cornea tissue removed during surgery revealed mild stromal textural alterations with alcianophilic deposits. Whole genome sequence data were generated for four affected individuals. No rare variants (MAF < 0.001) were identified in established corneal dystrophy genes. However, a novel heterozygous missense variant in exon 4 of SPARCL1, NM_004684: c.334G > A; p.(Glu112Lys), which is predicted to be damaging, segregated with disease. SPARC-like protein 1 (SPARCL1) is a secreted matricellular protein involved in cell migration, cell adhesion, tissue repair, and remodelling. Interestingly, SPARCL1 has been shown to regulate decorin. Heterozygous variants in DCN, encoding decorin, cause autosomal dominant congenital stromal corneal dystrophy, suggesting a common pathogenic pathway. Therefore, we performed immunohistochemistry to compare SPARCL1 and decorin localisation in corneal tissue from an affected family member and an unaffected control. Strikingly, the level of decorin was significantly decreased in the corneal stroma of the affected tissue, and SPARCL1 appeared to be retained in the epithelium. In summary, we describe a novel autosomal dominant corneal stromal dystrophy associated with a missense variant in SPARCL1, extending the phenotypic and genetic heterogeneity of inherited corneal disease.

19.
Am J Ophthalmol ; 258: 183-195, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37972748

RESUMEN

PURPOSE: To report the genetic etiology of Lisch epithelial corneal dystrophy (LECD). DESIGN: Multicenter cohort study. METHODS: A discovery cohort of 27 individuals with LECD from 17 families, including 7 affected members from the original LECD family, 6 patients from 2 new families and 14 simplex cases, was recruited. A cohort of 6 individuals carrying a pathogenic MCOLN1 (mucolipin 1) variant was reviewed for signs of LECD. Next-generation sequencing or targeted Sanger sequencing were used in all patients to identify pathogenic or likely pathogenic variants and penetrance of variants. RESULTS: Nine rare heterozygous MCOLN1 variants were identified in 23 of 27 affected individuals from 13 families. The truncating nature of 7 variants and functional testing of 1 missense variant indicated that they result in MCOLN1 haploinsufficiency. Importantly, in the homozygous and compound-heterozygous state, 4 of 9 LECD-associated variants cause the rare lysosomal storage disorder mucolipidosis IV (MLIV). Autosomal recessive MLIV is a systemic disease and comprises neurodegeneration as well as corneal opacity of infantile-onset with epithelial autofluorescent lysosomal inclusions. However, the 6 parents of 3 patients with MLIV confirmed to carry pathogenic MCOLN1 variants did not have the LECD phenotype, suggesting MCOLN1 haploinsufficiency may be associated with reduced penetrance and variable expressivity. CONCLUSIONS: MCOLN1 haploinsufficiency is the major cause of LECD. Based on the overlapping clinical features of corneal epithelial cells with autofluorescent inclusions reported in both LECD and MLIV, it is concluded that some carriers of MCOLN1 haploinsufficiency-causing variants present with LECD.


Asunto(s)
Distrofias Hereditarias de la Córnea , Mucolipidosis , Canales de Potencial de Receptor Transitorio , Humanos , Canales de Potencial de Receptor Transitorio/genética , Estudios de Cohortes , Mucolipidosis/diagnóstico , Mucolipidosis/genética , Mucolipidosis/patología , Distrofias Hereditarias de la Córnea/diagnóstico , Distrofias Hereditarias de la Córnea/genética
20.
Hum Mutat ; 34(3): 506-14, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23281133

RESUMEN

In one consanguineous family with retinitis pigmentosa (RP), a condition characterized by progressive visual loss due to retinal degeneration, homozygosity mapping, and candidate gene sequencing suggested a novel locus. Exome sequencing identified a homozygous frameshifting mutation, c.601delG, p.Lys203Argfs*28, in RP1L1 encoding RP 1-like1, a photoreceptor-specific protein. A screen of a further 285 unrelated individuals with autosomal recessive RP identified an additional proband, homozygous for a missense variant, c.1637G>C, p.Ser546Thr, in RP1L1. A distinct retinal disorder, occult macular dystrophy (OCMD) solely affects the central retinal cone photoreceptors and has previously been reported to be associated with variants in the same gene. The association between mutations in RP1L1 and the disorder OCMD was explored by screening a cohort of 28 unrelated individuals with the condition; 10 were found to harbor rare (minor allele frequency ≤0.5% in the 1,000 genomes dataset) heterozygous RP1L1 missense variants. Analysis of family members revealed many unaffected relatives harboring the same variant. Linkage analysis excluded the possibility of a recessive mode of inheritance, and sequencing of RP1, a photoreceptor protein that interacts with RP1L1, excluded a digenic mechanism involving this gene. These findings imply an important and diverse role for RP1L1 in human retinal physiology and disease.


Asunto(s)
Proteínas del Ojo/genética , Degeneración Macular/genética , Retina/patología , Retinitis Pigmentosa/genética , Adulto , Anciano , Mapeo Cromosómico , Clonación Molecular , Estudios de Cohortes , Consanguinidad , Exoma , Femenino , Genes Recesivos , Sitios Genéticos , Haplotipos , Heterocigoto , Homocigoto , Humanos , Masculino , Análisis por Micromatrices , Persona de Mediana Edad , Mutación Missense , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA