Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
PLoS Genet ; 20(5): e1011230, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38713708

RESUMO

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.


Assuntos
Distrofia Endotelial de Fuchs , Fator de Transcrição 4 , Expansão das Repetições de Trinucleotídeos , Humanos , Fator de Transcrição 4/genética , Fator de Transcrição 4/metabolismo , Expansão das Repetições de Trinucleotídeos/genética , Distrofia Endotelial de Fuchs/genética , Processamento Alternativo/genética , Transcriptoma/genética , Células Endoteliais/metabolismo , Endotélio Corneano/metabolismo , Endotélio Corneano/patologia , Masculino
2.
Am J Hum Genet ; 102(3): 447-459, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29499165

RESUMO

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.


Assuntos
Distrofias Hereditárias da Córnea/genética , Proteínas de Ligação a DNA/genética , Mutação/genética , Fatores de Transcrição/genética , Sequência de Bases , DNA Intergênico/genética , Endotélio Corneano/patologia , Família , Feminino , Loci Gênicos , Células HEK293 , Humanos , Íntrons/genética , Masculino , Modelos Genéticos , Linhagem , Regiões Promotoras Genéticas/genética , Transcrição Gênica , Sequenciamento Completo do Genoma
3.
Am J Hum Genet ; 102(4): 528-539, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29526280

RESUMO

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.


Assuntos
Distrofia Endotelial de Fuchs/genética , Predisposição Genética para Doença , Oligonucleotídeos Antissenso/farmacologia , Fator de Transcrição 4/genética , Expansão das Repetições de Trinucleotídeos/genética , Idoso , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Estudos de Coortes , Células Endoteliais/metabolismo , Endotélio Corneano/patologia , Feminino , Distrofia Endotelial de Fuchs/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Precursores de RNA/genética , Processamento Pós-Transcricional do RNA , Fatores de Processamento de RNA/metabolismo , RNA Mensageiro/metabolismo , Fatores de Risco
4.
Ophthalmology ; 128(11): 1516-1526, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33892046

RESUMO

PURPOSE: To examine the efficacy and safety of corneal cross-linking (CXL) for stabilization of progressive keratoconus. DESIGN: Observer-masked, randomized, controlled, parallel-group superiority trial. PARTICIPANTS: Sixty participants 10 to 16 years of age with progressive keratoconus, one eye of each deemed the study eye. METHODS: The study eye was randomized to either CXL plus standard care or standard care alone, with spectacle or contact lens correction as necessary for vision. MAIN OUTCOME MEASURES: The primary outcome was steep keratometry (K2) in the study eye as a measure of the steepness of the cornea at 18 months. Secondary outcomes included keratoconus progression defined as a 1.5-diopter (D) increase in K2, visual acuity, keratoconus apex corneal thickness, and quality of life. RESULTS: Of 60 participants, 30 were randomized to CXL and standard care groups. Of these, 30 patients in the CXL group and 28 patients in the standard care group were analyzed. Mean K2 in the study eye 18 months after randomization was 49.7 D (standard deviation [SD], 3.8 D) in the CXL group and 53.4 D (SD, 5.8 D) in the standard care group. The adjusted mean difference in K2 in the study eye was -3.0 D (95% confidence interval [CI], -4.9 to -1.1 D; P = 0.002), favoring CXL. Adjusted differences between groups in uncorrected and corrected vision favored eyes receiving CXL: -0.31 logarithm of the minimum angle of resolution (logMAR; 95% CI, -0.50 to -0.11 logMAR; P = 0.002) and -0.51 logMAR (95% CI, -1.37 to 0.35 logMAR; P = 0.002). Keratoconus progression in the study eye occurred in 2 patients (7%) randomized to CXL compared with 12 patients (43%) randomized to standard care. The unadjusted odds ratio suggests that on average, patients in the CXL arm had 90% (odds ratio, 0.1; 95% CI, 0.02-0.48; P = 0.004) lower odds of experiencing progression compared with those receiving standard care. CONCLUSIONS: CXL arrests progression of keratoconus in the majority of young patients. CXL should be considered as a first-line treatment in progressive disease. If the arrest of keratoconus progression induced by CXL is sustained in longer follow-up, particular benefit may be derived from avoiding a later requirement for contact lens wear or corneal transplantation.


Assuntos
Colágeno/uso terapêutico , Córnea/patologia , Ceratocone/tratamento farmacológico , Fotoquimioterapia/métodos , Refração Ocular/fisiologia , Riboflavina/uso terapêutico , Adolescente , Topografia da Córnea , Reagentes de Ligações Cruzadas/uso terapêutico , Feminino , Seguimentos , Humanos , Ceratocone/patologia , Masculino , Fármacos Fotossensibilizantes/uso terapêutico , Estudos Prospectivos , Resultado do Tratamento , Raios Ultravioleta
5.
Am J Hum Genet ; 99(6): 1338-1352, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27839872

RESUMO

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.


Assuntos
Segmento Anterior do Olho/anormalidades , Complemento C3/genética , Anormalidades do Olho/genética , Genes Recessivos/genética , Mutação , Inibidor da Tripsina Pancreática de Kazal/genética , alfa-Macroglobulinas/genética , Adolescente , Adulto , Sequência de Aminoácidos , Segmento Anterior do Olho/metabolismo , Criança , Pré-Escolar , Complemento C3/química , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inibidor da Tripsina Pancreática de Kazal/química , Adulto Jovem , alfa-Macroglobulinas/química
6.
Am J Hum Genet ; 98(1): 75-89, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26749309

RESUMO

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.


Assuntos
Alelos , Distrofias Hereditárias da Córnea/genética , Mutação , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Sequência de Bases , DNA , Feminino , Humanos , Masculino , Linhagem , Homologia de Sequência do Ácido Nucleico
7.
Genet Med ; 21(9): 2092-2102, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30733599

RESUMO

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.


Assuntos
Distrofia Endotelial de Fuchs/genética , Predisposição Genética para Doença , Fator de Transcrição 4/genética , Expansão das Repetições de Trinucleotídeos/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Sistemas CRISPR-Cas/genética , Feminino , Distrofia Endotelial de Fuchs/patologia , Genótipo , Humanos , Íntrons/genética , Masculino , Pessoa de Meia-Idade , Análise de Sequência de DNA , Imagem Individual de Molécula , Repetições de Trinucleotídeos/genética
8.
Exp Eye Res ; 182: 160-166, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30851240

RESUMO

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.


Assuntos
Distrofias Hereditárias da Córnea/genética , DNA/genética , Mutação , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Adolescente , Adulto , Idoso , Sequência de Bases , Criança , Pré-Escolar , Distrofias Hereditárias da Córnea/diagnóstico , Distrofias Hereditárias da Córnea/metabolismo , Análise Mutacional de DNA , Éxons , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Pessoa de Meia-Idade , Linhagem , Deleção de Sequência , Adulto Jovem , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , Dedos de Zinco
9.
BMC Ophthalmol ; 18(1): 250, 2018 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-30223810

RESUMO

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.


Assuntos
Distrofias Hereditárias da Córnea/genética , DNA/genética , Dimetilaliltranstransferase/genética , Mutação , Adulto , Criança , Distrofias Hereditárias da Córnea/diagnóstico , Distrofias Hereditárias da Córnea/metabolismo , Análise Mutacional de DNA , Dimetilaliltranstransferase/metabolismo , Feminino , Humanos , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Linhagem , Fenótipo , Tomografia de Coerência Óptica/métodos
10.
Hum Mutat ; 36(4): 463-73, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25676728

RESUMO

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.


Assuntos
Autoantígenos/genética , Distrofias Hereditárias da Córnea/diagnóstico , Distrofias Hereditárias da Córnea/genética , Epitélio Corneano/patologia , Estudos de Associação Genética , Mutação , Colágenos não Fibrilares/genética , Adolescente , Adulto , Idade de Início , Idoso , Idoso de 80 Anos ou mais , Autoantígenos/metabolismo , Criança , Feminino , Expressão Gênica , Variação Genética , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Colágenos não Fibrilares/metabolismo , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único , Splicing de RNA , Adulto Jovem , Colágeno Tipo XVII
11.
Ann Hum Genet ; 79(1): 1-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25441224

RESUMO

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.


Assuntos
Distrofias Hereditárias da Córnea/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Adolescente , Adulto , Criança , Códon sem Sentido , Análise Mutacional de DNA , Feminino , Mutação da Fase de Leitura , Humanos , Masculino , Linhagem , Fenótipo , Sítios de Splice de RNA/genética , Deleção de Sequência , Adulto Jovem , Homeobox 1 de Ligação a E-box em Dedo de Zinco
12.
Am J Hum Genet ; 90(2): 247-59, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22284829

RESUMO

X-linked megalocornea (MGC1) is an ocular anterior segment disorder characterized by an increased cornea diameter and deep anterior chamber evident at birth and later onset of mosaic corneal degeneration (shagreen), arcus juvenilis, and presenile cataracts. We identified copy-number variation, frameshift, missense, splice-site and nonsense mutations in the Chordin-like 1 gene (CHRDL1) on Xq23 as the cause of the condition in seven MGC1 families. CHRDL1 encodes ventroptin, a bone morphogenic protein antagonist with a proposed role in specification of topographic retinotectal projections. Electrophysiological evaluation revealed mild generalized cone system dysfunction and, in one patient, an interhemispheric asymmetry in visual evoked potentials. We show that CHRDL1 is expressed in the developing human cornea and anterior segment in addition to the retina. We explored the impact of loss of ventroptin function on brain function and morphology in vivo. CHRDL1 is differentially expressed in the human fetal brain, and there is high expression in cerebellum and neocortex. We show that MGC1 patients have a superior cognitive ability despite a striking focal loss of myelination of white matter. Our findings reveal an unexpected requirement for ventroptin during anterior segment development and the consequences of a lack of function in the retina and brain.


Assuntos
Segmento Anterior do Olho/embriologia , Córnea/anormalidades , Anormalidades do Olho/genética , Proteínas do Olho/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Mutação , Proteínas do Tecido Nervoso/genética , Adulto , Segmento Anterior do Olho/anormalidades , Sequência de Bases , Encéfalo/patologia , Paralisia Cerebral/genética , Paralisia Cerebral/metabolismo , Doenças da Córnea/genética , Doenças da Córnea/metabolismo , Variações do Número de Cópias de DNA/genética , Anormalidades do Olho/complicações , Anormalidades do Olho/embriologia , Proteínas do Olho/biossíntese , Feminino , Genes Ligados ao Cromossomo X , Doenças Genéticas Ligadas ao Cromossomo X/complicações , Doenças Genéticas Ligadas ao Cromossomo X/embriologia , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Masculino , Megalencefalia/genética , Megalencefalia/metabolismo , Pessoa de Meia-Idade , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/biossíntese , Linhagem , Fenótipo , Locos de Características Quantitativas , Retina/anormalidades , Retina/embriologia , Adulto Jovem
13.
Exp Eye Res ; 138: 70-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26142953

RESUMO

Human limbal epithelial stem cells (LESCs) are essential for the maintenance of the corneal epithelium of the ocular surface. LESCs are located within limbal crypts between the palisades of Vogt in the limbus; the interface between the peripheral cornea and conjunctiva. The limbal crypts have been proposed as a LESC niche owing to their support of epithelial cells, which can form holoclone colonies in vitro. Closely associated with the limbal crypts is a concentrated population of melanocytes. The anatomical location and close proximity to putative LESC suggests that melanocytes might play a role in maintenance of these stem cells in the niche. The aim of this study was to assess the ability of human limbal melanocytes (hLM) to support the expansion of human limbal epithelial cells (LECs) in vitro as an indicator of functional cell-cell interaction. After observing that hLM co-localize with clusters of compact epithelial cells in the native limbal crypts, hLM were isolated from crypt-rich cadaveric limbal biopsies and used as feeders for the culture of LECs. Interestingly, LECs grown on mitotically active hLM were able to generate large epithelial colonies that contained small and compact cells with morphological stem cell characteristics. Immunocytochemistry revealed that LECs expanded on hLM were positive for the expression of the putative stem cell markers CK15, Bmi-1 and p63α and negative for the marker of terminal cell differentiation CK3. LECs and hLM were finally co-cultured on RAFT (real architecture for 3D tissue) collagen tissue equivalents. In 3D co-cultures, hLM promoted multi-layering of the epithelial sheet in which basal cells were maintained in an undifferentiated state. Taken together, these observations suggest melanocytes could play an important role in the maintenance of LESCs in the native human limbal stem cell niche.


Assuntos
Epitélio Corneano/citologia , Limbo da Córnea/citologia , Melanócitos/fisiologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/citologia , Biomarcadores , Comunicação Celular/fisiologia , Contagem de Células , Técnicas de Cocultura , Epitélio Corneano/metabolismo , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Limbo da Córnea/metabolismo , Microscopia Eletrônica de Transmissão , Células-Tronco/metabolismo , Doadores de Tecidos , Engenharia Tecidual
14.
Eye (Lond) ; 38(9): 1681-1686, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38409307

RESUMO

OBJECTIVE: To define how estimates of keratoconus progression following collagen cross-linking (CXL) vary according to the parameter selected to measure corneal shape. MATERIALS AND METHODS: We estimated progression following CXL in 1677 eyes. We compared standard definitions of keratoconus progression based on published thresholds for Kmax, front K2, or back K2, or progression of any two of these three parameters, with the option of an increased threshold for Kmax values ≥ 55D. As corneal thickness reduces unpredictably after CXL, it was excluded from the principal analysis. We then repeated the analysis using novel adaptive estimates of progression for Kmax, front K2, or back K2, developed separately using 6463 paired readings from keratoconus eyes, with a variation of the Bland-Altman method to determine the 95% regression-based limits of agreement (LoA). We created Kaplan-Meier survival plots for both standard and adaptive thresholds. The primary outcome was progression five years after a baseline visit 9-15 months following CXL. RESULTS: Progression rates were 8% with a standard (≥ 1.5D) threshold for K2 or 6% with the static multi-parameter definition. With a ≥ 1D threshold for Kmax, the progression was significantly higher at 29%. With adaptive Kmax or K2, the progression rates were similar (20%) but less than with the adaptive multi-parameter method (22%). CONCLUSIONS: Estimates of keratoconus progression following CXL vary widely according to the reference criteria. Using adaptive thresholds (LoA) to define the repeatability of keratometry gives estimates for progression that are markedly higher than with the standard multi-parameter method.


Assuntos
Colágeno , Córnea , Topografia da Córnea , Reagentes de Ligações Cruzadas , Progressão da Doença , Ceratocone , Fármacos Fotossensibilizantes , Riboflavina , Ceratocone/tratamento farmacológico , Ceratocone/diagnóstico , Ceratocone/fisiopatologia , Humanos , Colágeno/metabolismo , Reagentes de Ligações Cruzadas/uso terapêutico , Masculino , Feminino , Adulto , Fármacos Fotossensibilizantes/uso terapêutico , Riboflavina/uso terapêutico , Córnea/patologia , Raios Ultravioleta , Acuidade Visual/fisiologia , Adulto Jovem , Fotoquimioterapia/métodos , Paquimetria Corneana , Adolescente , Substância Própria/metabolismo , Substância Própria/patologia
15.
Am J Ophthalmol ; 258: 183-195, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37972748

RESUMO

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.


Assuntos
Distrofias Hereditárias da Córnea , Mucolipidoses , Canais de Potencial de Receptor Transitório , Humanos , Canais de Potencial de Receptor Transitório/genética , Estudos de Coortes , Mucolipidoses/diagnóstico , Mucolipidoses/genética , Mucolipidoses/patologia , Distrofias Hereditárias da Córnea/diagnóstico , Distrofias Hereditárias da Córnea/genética
16.
Acta Ophthalmol ; 101(6): 679-686, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36883248

RESUMO

PURPOSE: To characterise the phenotype and genotype of concurrent keratoconus and Fuchs endothelial corneal dystrophy (KC + FECD). METHODS: We recruited 20 patients with concurrent KC + FECD for a retrospective observational case series from the United Kingdom and the Czech Republic. We compared eight parameters of corneal shape (Pentacam, Oculus) with two groups of age-matched controls who had either isolated keratoconus (KC) or isolated FECD. We genotyped probands for an intronic triplet TCF4 repeat expansion (CTG18.1) and the ZEB1 variant c.1920G >T p.(Gln640His). RESULTS: The median age at diagnosis of patients with KC + FECD was 54 (interquartile range 46 to 66) years, with no evidence of KC progression (median follow-up 84 months, range 12 to 120 months). The mean (standard deviation (SD)) of the minimum corneal thickness, 493 (62.7) µm, was greater than eyes with KC, 458 (51.1) µm, but less than eyes with FECD, 590 (55.6) µm. Seven other parameters of corneal shape were more like KC than FECD. Seven (35%) probands with KC + FECD had a TCF4 repeat expansion of ≥50 compared to five controls with isolated FECD. The average of the largest TCF4 expansion in cases with KC + FECD (46 repeats, SD 36 repeats) was similar to the age-matched controls with isolated FECD (36 repeats, SD 28 repeats; p = 0.299). No patient with KC + FECD harboured the ZEB1 variant. CONCLUSIONS: The KC + FECD phenotype is consistent with KC but with superimposed stromal swelling from endothelial disease. The proportion of cases with a TCF4 expansion is similar in concurrent KC + FECD and age-matched controls with isolated FECD.


Assuntos
Distrofia Endotelial de Fuchs , Ceratocone , Humanos , Distrofia Endotelial de Fuchs/complicações , Distrofia Endotelial de Fuchs/diagnóstico , Distrofia Endotelial de Fuchs/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fator de Transcrição 4/genética , Estudos Retrospectivos , Ceratocone/complicações , Ceratocone/diagnóstico , Ceratocone/genética , Fatores de Transcrição/genética , Genótipo , Fenótipo
17.
Ophthalmology ; 119(9): 1766-72, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22683059

RESUMO

OBJECTIVE: We sought to describe the clinical presentation, effect on visual acuity, impact on school attendance, and access to appropriate eye care in children with vernal keratoconjunctivitis (VKC) in Rwanda (Central Africa). DESIGN: Case-control study nested within a cross-sectional survey. PARTICIPANTS: We examined 3041 children; 121 had VKC. METHODS: Primary schools were randomly selected and children were interviewed using a questionnaire on VKC-related symptoms. Data on health-seeking behavior and school attendance were recorded. Children received a full eye examination, including visual acuity using a LogMar E Chart. MAIN OUTCOME MEASURES: Description of the clinical findings, unaided visual acuity, prior attendance for medical eye care, and the impact of VKC on school attendance. RESULTS: Of the 121 children with VKC, 119 (98.4%) had only limbal disease. Ocular itching (n = 101; 83.5%) was the predominant symptom and this was seasonal in 66 children (65.4%), constant but with variable intensity in 18 (17.8%), and constant with constant intensity in 17 children (16.8%). Children with VKC were >6 times more likely to have corneal astigmatism >2 diopters in their worse eye (odds ratio [OR], 6.31; 95% confidence interval [CI], 3.26-12.26; P<0.001) than controls. Eight affected eyes had astigmatism >4 diopters or irregular astigmatism incompatible with autokeratometry. Although 4 eyes (1.7%) had uncorrected low vision from VKC-induced corneal astigmatism or keratoconus, only 1 child was visually impaired in both eyes. School nonattendance for an ocular reason during the last 3 months was 5 times more likely in children with VKC (n = 44; 36.4%) than among those without (n = 297; 10.2%; OR, 5.04; 95% CI, 3.40-7.47; P < 0.001). Repeating a school year or having ever dropped out of school was not more common among children with VKC than those without. Medical eye care had been sought by 54 (44.6%) children with VKC. CONCLUSIONS: This survey of prevalence and treatment of VKC in an African community adds to the argument for better primary eye care, including a safe topical medication. Long-term follow-up of this cohort is required to ascertain the overall risk of sight-threatening complications.


Assuntos
Absenteísmo , Conjuntivite Alérgica/epidemiologia , Acessibilidade aos Serviços de Saúde/estatística & dados numéricos , Instituições Acadêmicas/estatística & dados numéricos , Estudantes/estatística & dados numéricos , Adolescente , Estudos de Casos e Controles , Criança , Conjuntivite Alérgica/diagnóstico , Estudos Transversais , Feminino , Inquéritos Epidemiológicos , Humanos , Masculino , Vigilância da População , Prevalência , Ruanda/epidemiologia , Inquéritos e Questionários , Acuidade Visual/fisiologia
18.
Int Ophthalmol ; 32(5): 471-3, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22581322

RESUMO

We describe a patient with severe atopic dermatitis and keratoconjunctivitis, who was prescribed both systemic and topical ciclosporin and who developed ocular surface squamous neoplasia (OSSN). To provide additional information on the incidence of the association between topical ciclosporin use and OSSN, we counted the total prescriptions for topical ciclosporin issued in our hospital between 2003 and 2006 (804 patients) and, for the same period, we reviewed the records of patients with OSSN for a history of treatment with ciclosporin or a history of atopy. No other case of OSSN in a patient with a history of topical ciclosporin use was identified, which makes it difficult to implicate topical ciclosporin as the causative factor.


Assuntos
Carcinoma de Células Escamosas/etiologia , Túnica Conjuntiva/patologia , Neoplasias da Túnica Conjuntiva/etiologia , Hospedeiro Imunocomprometido , Ceratoconjuntivite/complicações , Carcinoma de Células Escamosas/diagnóstico , Neoplasias da Túnica Conjuntiva/diagnóstico , Diagnóstico Diferencial , Feminino , Humanos , Ceratoconjuntivite/diagnóstico , Ceratoconjuntivite/imunologia , Pessoa de Meia-Idade , Fotomicrografia
19.
J Clin Med ; 11(17)2022 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-36079096

RESUMO

We report the phenotype of a 15-year-old female patient with anterior segment dysgenesis (ASD) caused by a novel heterozygous loss-of-function FOXC1 variant. The proband underwent an ophthalmic examination as well as a molecular genetic investigation comprising exome sequencing, a single nucleotide polymorphism array to access copy number and Sanger sequencing to exclude non-coding causal variants. There was bilateral mild iris hypoplasia with pupil deformation and iridocorneal adhesions. In addition to these features of ASD, the corneas were flat, with mean keratometry readings of 38.8 diopters in the right eye and 39.5 diopters in the left eye. There was a snail track lesion of the left cornea at the level of the Descemet membrane. The central corneal endothelial cell density was reduced bilaterally at 1964 and 1373 cells/mm2 in the right and left eyes, respectively. Molecular genetic analysis revealed that the proband was a carrier of a novel heterozygous frameshifting variant in FOXC1, c.605del p.(Pro202Argfs*113). Neither parent had this change, suggesting a de novo origin which was supported by paternity testing. We found no possibly pathogenic variants in the other genes associated with posterior corneal dystrophies or ASD. Further studies are warranted to verify whether there is a true association between snail track lesions, corneal flattening, and pathogenic variants in FOXC1.

20.
Am J Ophthalmol ; 240: 321-329, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35469790

RESUMO

PURPOSE: To generate a prognostic model to predict keratoconus progression to corneal crosslinking (CXL). DESIGN: Retrospective cohort study. METHODS: We recruited 5025 patients (9341 eyes) with early keratoconus between January 2011 and November 2020. Genetic data from 926 patients were available. We investigated both keratometry or CXL as end points for progression and used the Royston-Parmar method on the proportional hazards scale to generate a prognostic model. We calculated hazard ratios (HRs) for each significant covariate, with explained variation and discrimination, and performed internal-external cross validation by geographic regions. RESULTS: After exclusions, model fitting comprised 8701 eyes, of which 3232 underwent CXL. For early keratoconus, CXL provided a more robust prognostic model than keratometric progression. The final model explained 33% of the variation in time to event: age HR (95% CI) 0.9 (0.90-0.91), maximum anterior keratometry 1.08 (1.07-1.09), and minimum corneal thickness 0.95 (0.93-0.96) as significant covariates. Single-nucleotide polymorphisms (SNPs) associated with keratoconus (n=28) did not significantly contribute to the model. The predicted time-to-event curves closely followed the observed curves during internal-external validation. Differences in discrimination between geographic regions was low, suggesting the model maintained its predictive ability. CONCLUSIONS: A prognostic model to predict keratoconus progression could aid patient empowerment, triage, and service provision. Age at presentation is the most significant predictor of progression risk. Candidate SNPs associated with keratoconus do not contribute to progression risk.


Assuntos
Ceratocone , Fotoquimioterapia , Colágeno/uso terapêutico , Topografia da Córnea , Demografia , Humanos , Ceratocone/diagnóstico , Ceratocone/tratamento farmacológico , Ceratocone/genética , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/uso terapêutico , Estudos Retrospectivos , Riboflavina/uso terapêutico , Raios Ultravioleta , Acuidade Visual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA