Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cureus ; 15(7): e42401, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37621826

ABSTRACT

Background Silver possesses cytotoxic properties against many microorganisms and is regularly used in wound care. Current evidence supporting the use of one type of silver-containing wound dressing (SCWD) is insufficient. Materials and methods To examine the ability of selected SCWDs to inhibit the growth of two strains of bacteria (Escherichia coli and Staphylococcus aureus) commonly found in wounds, an in vitro wound model was used. Bacteria were applied to the surface of nutrient agar, and a piece of each SCWD was applied to the bacteria. The plates were incubated at 37°C overnight. The zone of inhibition (ZI) around each SCWD was measured in cm2. Results The mean ZI for Acticoat Flex-3 on E. coli was 1.59 ± 0.15 cm2, which was significantly greater than that observed for Aquacel Ag (p<0.001), Mepilex Ag (p<0.0001), Mepitel Ag (p<0.001), Optifoam (p<0.0001), and Tegaderm Alginate Ag (p<0.01), but statistically indistinguishable from Maxorb II Ag. The mean ZI on S. aureus was 1.21 ± 0.16 cm2, which was greater than Aquacel Ag (p<0.05), Mepilex (p<0.0001), Optifoam (p<0.0001), and Tegaderm Alginate Ag (p<0.05), but statistically indistinguishable from Maxorb II Ag or Mepitel Ag. Conclusion Of the SCWDs tested, Acticoat Flex-3 demonstrated the most robust antimicrobial effect. Herein, we show that Acticoat Flex-3 may provide the most wound protection against bacterial infection. In conclusion, these data provide clinicians with additional independent evidence to inform their clinical practice on the use of specific wound dressings.

2.
Ophthalmic Genet ; 42(4): 486-492, 2021 08.
Article in English | MEDLINE | ID: mdl-34003075

ABSTRACT

BACKGROUND: Concomitant corneal ectasia and posterior lamellar corneal opacification is rare, and the genetic relationship between these two conditions is unclear. We report the genetic and clinical characterization of this phenotype in three unrelated individuals. MATERIALS AND METHODS: One previously reported affected individual and two unreported, unrelated, affected individuals were recruited for the study. Subjects and unaffected relatives underwent slit lamp examination, refraction, and multi-modal imaging. Saliva samples were obtained from two of the three affected individuals, from which DNA was extracted. Sanger sequencing was performed to identify mutations in genes associated with posterior amorphous corneal dystrophy (PACD), brittle cornea syndrome (BCS), and posterior polymorphous corneal dystrophy (PPCD), while copy number variation (CNV) analysis was used to identify CNV in the PACD locus. RESULTS: Affected individuals demonstrated bilateral corneal steepening, stromal thinning and lamellar posterior corneal opacification. Corneal topography and tomography revealed conical or globular corneal steepening and decreased thickness. Anterior segment optical coherence tomography demonstrated hyperreflectivity of the posterior stroma in each of the affected individuals. Genetic testing did not detect a heterozygous deletion involving the PACD locus on chromosome 12 or a pathogenic mutation in the genes associated with BCS or PPCD. CONCLUSIONS: Corneal ectasia may be associated with posterior lamellar stromal opacification that appears consistent with PACD. However, genetic testing for PACD as well as BCS and PPCD in affected individuals fails to reveal pathogenic deletions or mutations, indicating that other genetic factors are involved.


Subject(s)
Cornea/pathology , Corneal Opacity/diagnosis , Corneal Stroma/pathology , Keratoconus/diagnosis , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/genetics , Corneal Topography , DNA Copy Number Variations , Dilatation, Pathologic , Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Female , Humans , Infant, Newborn , Joint Instability/congenital , Joint Instability/diagnosis , Joint Instability/genetics , Male , Mutation , Skin Abnormalities/diagnosis , Skin Abnormalities/genetics , Slit Lamp Microscopy , Young Adult
3.
Cornea ; 39(11): 1442-1445, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32482962

ABSTRACT

PURPOSE: To investigate the presence of pre-Descemet corneal dystrophy (PDCD) in association with X-linked ichthyosis (XLI) in an 11-year-old boy using multimodal imaging and genetic analysis. METHODS: Corneal opacities were examined and imaged with slit-lamp biomicroscopy, anterior segment optical coherence tomography, noncontact specular microscopy, and in vivo confocal microscopy. Cytogenomic array analysis was performed using genomic DNA isolated from the patient. RESULTS: Corneal opacities characteristic of PDCD located in the posterior corneal stroma just anterior to Descemet membrane were identified by slit-lamp biomicroscopy. A pre-Descemet hyper-reflective line, consistent with these opacities, was seen with anterior segment optical coherence tomography. Scheimpflug tomography revealed a bimodal peak light scattering. In vivo confocal microscopy findings were unremarkable. Copy number analysis identified a 4389 kbp hemizygous deletion on the X chromosome (chr. X: 6,540,898-8,167,604), resulting in the deletion of 4 genes, including the known locus of XLI, the STS gene. CONCLUSIONS: This report demonstrates that PDCD-associated XLI may present in children and that the diagnosis may be confirmed through multimodal imaging in conjunction with genetic analysis.


Subject(s)
Corneal Dystrophies, Hereditary/diagnosis , Ichthyosis, X-Linked/diagnosis , Microscopy, Confocal/methods , Multimodal Imaging , Slit Lamp Microscopy/methods , Steryl-Sulfatase/genetics , Tomography, Optical Coherence/methods , Child , Corneal Dystrophies, Hereditary/genetics , Corneal Stroma/pathology , DNA/genetics , Descemet Membrane/pathology , Humans , Ichthyosis, X-Linked/genetics , Male , Steryl-Sulfatase/metabolism
4.
Sci Rep ; 10(1): 7402, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366916

ABSTRACT

The advent of cell culture-based methods for the establishment and expansion of human corneal endothelial cells (CEnC) has provided a source of transplantable corneal endothelium, with a significant potential to challenge the one donor-one recipient paradigm. However, concerns over cell identity remain, and a comprehensive characterization of the cultured CEnC across serial passages has not been performed. To this end, we compared two established CEnC culture methods by assessing the transcriptomic changes that occur during in vitro expansion. In confluent monolayers, low mitogenic culture conditions preserved corneal endothelial cell state identity better than culture in high mitogenic conditions. Expansion by continuous passaging induced replicative cell senescence. Transcriptomic analysis of the senescent phenotype identified a cell senescence signature distinct for CEnC. We identified activation of both classic and new cell signaling pathways that may be targeted to prevent senescence, a significant barrier to realizing the potential clinical utility of in vitro expansion.


Subject(s)
Cell Culture Techniques/methods , Endothelium, Corneal/cytology , Adolescent , Adult , Cell Movement , Cell Proliferation , Cellular Senescence , Child , Child, Preschool , Computational Biology , Corneal Transplantation , Female , Humans , Male , Phenotype , Signal Transduction , Transcriptome , Young Adult
5.
PLoS One ; 14(6): e0218279, 2019.
Article in English | MEDLINE | ID: mdl-31194824

ABSTRACT

The zinc finger e-box binding homeobox 1 (ZEB1) transcription factor is a master regulator of the epithelial to mesenchymal transition (EMT), and of the reverse mesenchymal to epithelial transition (MET) processes. ZEB1 plays an integral role in mediating cell state transitions during cell lineage specification, wound healing and disease. EMT/MET are characterized by distinct changes in molecular and cellular phenotype that are generally context-independent. Posterior polymorphous corneal dystrophy (PPCD), associated with ZEB1 insufficiency, provides a new biological context in which to understand and evaluate the classic EMT/MET paradigm. PPCD is characterized by a cadherin-switch and transition to an epithelial-like transcriptomic and cellular phenotype, which we study in a cell-based model of PPCD generated using CRISPR-Cas9-mediated ZEB1 knockout in corneal endothelial cells (CEnCs). Transcriptomic and functional studies support the hypothesis that CEnC undergo a MET-like transition in PPCD, termed endothelial to epithelial transition (EnET), and lead to the conclusion that EnET may be considered a corollary to the classic EMT/MET paradigm.


Subject(s)
Endothelium, Corneal/metabolism , Epithelial-Mesenchymal Transition/physiology , Zinc Finger E-box-Binding Homeobox 1/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation , Cornea/metabolism , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation/genetics , Homeodomain Proteins/genetics , Humans , Transcription Factors/metabolism , Transcriptome , Zinc Finger E-box-Binding Homeobox 1/genetics
6.
Ophthalmic Genet ; 39(4): 419-424, 2018 08.
Article in English | MEDLINE | ID: mdl-29671669

ABSTRACT

PURPOSE: To present the clinical and cytogenetic features of a previously unreported family with posterior amorphous corneal dystrophy (PACD) associated with a heterozygous deletion of the small leucine-rich proteoglycan (SRLP) genes on chromosome 12. METHODS: Clinical characterization was performed using slit lamp biomicroscopic and optical coherence tomography (OCT) imaging. Genomic DNA was collected from affected and unaffected family members, and a cytogenomic array was used to identify copy number variations (CNV) present in the PACD locus. RESULTS: Three members of a Guatemalan family presented with clinical characteristics consistent with PACD: bilateral posterior stromal lamellar opacification, decreased corneal curvature, and iridocorneal adhesions. OCT imaging demonstrated decreased corneal thickness and hyperreflectivity of the posterior third of the corneal stroma. CNV analysis confirmed the presumed clinical diagnosis of PACD by revealing a 0.304 Mb heterozygous deletion in the PACD locus on chromosome 12 that included the four SLRP genes (KERA, LUM, DCN, and EPYC) deleted in each of the PACD families in which CNV analysis has been reported. CONCLUSIONS: This is the first report of the OCT appearance of PACD and the second confirmation of a heterozygous deletion of chromosome 12q21.33 as the cause of PACD, highlighting the utility of array-based cytogenomics to confirm the suspected clinical diagnosis of PACD. As the smallest previously reported pathogenic deletion was 0.701 Mb, the 0.304-Mb deletion we report is the smallest identified to date and reduces the size of the PACD locus to 0.275 Mb.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Corneal Dystrophies, Hereditary/genetics , DNA Copy Number Variations , Sequence Deletion , Small Leucine-Rich Proteoglycans/genetics , Adolescent , Child, Preschool , Corneal Dystrophies, Hereditary/diagnostic imaging , Corneal Topography , Female , Heterozygote , Humans , Male , Middle Aged , Mutation , Pedigree , Slit Lamp Microscopy , Tomography, Optical Coherence
7.
Mol Vis ; 23: 740-752, 2017.
Article in English | MEDLINE | ID: mdl-29046608

ABSTRACT

PURPOSE: To investigate the functional role that the zinc e-box binding homeobox 1 (ZEB1) gene, which underlies the genetic basis of posterior polymorphous corneal dystrophy 3 (PPCD3), plays in corneal endothelial cell proliferation, apoptosis, migration, and barrier function. METHODS: A human corneal endothelial cell line (HCEnC-21T) was transfected with siRNA targeting ZEB1 mRNA. Cell proliferation, apoptosis, migration, and barrier assays were performed: Cell proliferation was assessed with cell counting using a hemocytometer; cell apoptosis, induced by either ultraviolet C (UVC) radiation or doxorubicin treatment, was quantified by measuring cleaved caspase 3 (cCASP3) protein levels; and cell migration and barrier function were monitored with electric cell-substrate impedance sensing (ECIS). RESULTS: ZEB1 knockdown in HCEnC-21T cells transfected with siRNA targeting ZEB1 did not result in a significant difference in cell proliferation when compared with control. Although knockdown of ZEB1 in HCEnC-21T cells sensitized the cells to UV-induced apoptosis, ZEB1 knockdown did not alter the cells' susceptibility to doxorubicin-induced apoptosis, as measured with cCASP3 protein levels, compared with controls. Similarly, no difference was observed in cell migration following ZEB1 knockdown. However, cell barrier function increased significantly following ZEB1 knockdown. CONCLUSIONS: The corneal endothelium in PPCD3 is characterized by morphologic, anatomic, and molecular features that are more consistent with an epithelial-like rather than an endothelial-like phenotype. Although these characteristics have been well documented, we demonstrate for the first time that susceptibility to UV-induced apoptosis and cell barrier function are significantly altered in the setting of reduced ZEB1. The significance of an altered cellular response to apoptotic stimuli and increased cell barrier function in the pathobiology of PPCD remains to be fully elucidated.


Subject(s)
Corneal Dystrophies, Hereditary/physiopathology , Endothelium, Corneal/physiology , Gene Expression Regulation/physiology , Zinc Finger E-box-Binding Homeobox 1/genetics , Antibiotics, Antineoplastic/toxicity , Apoptosis/physiology , Blotting, Western , Caspase 3/metabolism , Cell Line , Cell Movement/physiology , Cell Proliferation/physiology , Doxorubicin/toxicity , Electric Impedance , Endothelium, Corneal/radiation effects , Gene Knockdown Techniques , Humans , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Transfection , Ultraviolet Rays/adverse effects
8.
Invest Ophthalmol Vis Sci ; 58(7): 3202-3214, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28654985

ABSTRACT

Purpose: To investigate the molecular basis of posterior polymorphous corneal dystrophy (PPCD) by examining the PPCD transcriptome and the effect of decreased ZEB1 expression on corneal endothelial cell (CEnC) gene expression. Methods: Next-generation RNA sequencing (RNA-seq) analyses of corneal endothelium from two PPCD-affected individuals (one with PPCD3 and one of unknown genetic cause) compared with two age-matched controls, and primary human CEnC (pHCEnC) transfected with siRNA-mediated ZEB1 knockdown. The expression of selected differentially expressed genes was validated by quantitative polymerase chain reaction (qPCR) and/or assessed by in situ hybridization in the corneal endothelium of four independent cases of PPCD (one with PPCD3 and three of unknown genetic cause). Results: Expression of 16% and 46% of the 104 protein-coding genes specific to ex vivo corneal endothelium was lost in the endothelium of two individuals with PPCD. Thirty-two genes associated with ZEB1 and 3 genes (BMP4, CCND1, ZEB1) associated with OVOL2 were differentially expressed in the same direction in both individuals with PPCD. Immunohistochemistry staining and RNA-seq analyses demonstrated variable expression of type IV collagens in PPCD corneas. Decreasing ZEB1 expression in pHCEnC altered expression of 711 protein-coding genes, many of which are associated with canonical pathways regulating various cellular processes. Conclusions: Identification of the altered transcriptome in PPCD and in a cell-based model of PPCD provided insight into the molecular alterations characterizing PPCD. Further study of the differentially expressed genes associated with ZEB1 and OVOL2 is expected to identify candidate genes for individuals with PPCD and without a ZEB1 or OVOL2 mutation.


Subject(s)
Collagen/metabolism , Corneal Dystrophies, Hereditary/genetics , Transcriptome , Adolescent , Adult , Case-Control Studies , Collagen Type IV/metabolism , Corneal Dystrophies, Hereditary/metabolism , Endothelial Cells/metabolism , Endothelium, Corneal/metabolism , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Polymerase Chain Reaction , Young Adult , Zinc Finger E-box-Binding Homeobox 1/metabolism
9.
PLoS One ; 12(1): e0169215, 2017.
Article in English | MEDLINE | ID: mdl-28046031

ABSTRACT

PURPOSE: To identify the genetic basis of posterior polymorphous corneal dystrophy (PPCD) in families mapped to the PPCD1 locus and in affected individuals without ZEB1 coding region mutations. METHODS: The promoter, 5' UTR, and coding regions of OVOL2 was screened in the PPCD family in which linkage analysis established the PPCD1 locus and in 26 PPCD probands who did not harbor a ZEB1 mutation. Copy number variation (CNV) analysis in the PPCD1 and PPCD3 intervals was performed on DNA samples from eight probands using aCGH. Luciferase reporter assays were performed in human corneal endothelial cells to determine the impact of the identified potentially pathogenic variants on OVOL2 promoter activity. RESULTS: OVOL2 mutation analysis in the first PPCD1-linked family demonstrated segregation of the c.-307T>C variant with the affected phenotype. In the other 26 probands screened, one heterozygous coding region variant and five promoter region heterozygous variants were identified, though none are likely pathogenic based on allele frequency. Array CGH in the PPCD1 and PPCD3 loci excluded the presence of CNV involving either OVOL2 or ZEB1, respectively. The c.-307T>C variant demonstrated increased promoter activity in corneal endothelial cells when compared to the wild-type sequence as has been demonstrated previously in another cell type. CONCLUSIONS: Previously identified as the cause of PPCD1, the OVOL2 promoter variant c.-307T>C was herein identified in the original family that established the PPCD1 locus. However, the failure to identify presumed pathogenic coding or non-coding OVOL2 or ZEB1 variants, or CNV involving the PPCD1 and PPCD3 loci in 26 other PPCD probands suggests that other genetic loci may be involved in the pathogenesis of PPCD.


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Mutation/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , 5' Untranslated Regions/genetics , Cell Line , Chromosome Segregation/genetics , Chromosomes, Human, Pair 20/genetics , DNA Copy Number Variations/genetics , Family , Female , Genetic Loci , Humans , Male , Pedigree , Reproducibility of Results , Zinc Finger E-box-Binding Homeobox 1/genetics
10.
Cornea ; 36(2): 210-216, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28060069

ABSTRACT

PURPOSE: To describe 2 unrelated families with multiple members demonstrating a less commonly recognized vortex pattern of corneal deposits confirmed to be granular corneal dystrophy type 1 (GCD1) after identification of the p.(Arg555Trp) mutation in the transforming growth factor ß-induced gene (TGFBI). METHODS: A slit-lamp examination was performed on individuals from 2 families, one of Mexican descent and a second of Italian descent. After DNA extraction from affected individuals and their unaffected relatives, TGFBI screening was performed. RESULTS: Eight of 20 individuals in the Mexican family and 20 of 55 in the Italian family demonstrated corneal stromal opacities. Seven of the 8 affected individuals in the Mexican family and 4 of the 20 affected individuals in the Italian family demonstrated a phenotype characterized by a "sea fan" or vortex pattern of superficial stromal corneal deposits originating from the inferior aspect of the cornea. Screening of TGFBI in both families revealed a heterozygous missense mutation [p.(Arg555Trp)] in exon 12, confirming the diagnosis of GCD1. CONCLUSIONS: Our findings demonstrate that GCD1 may present with a vortex pattern of anterior stromal deposits. Although this pattern of dystrophic deposits is not recognized by clinicians as a typical phenotype of GCD1, it is consistent with the production of the majority of the TGFBI protein by the corneal epithelium.


Subject(s)
Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/genetics , Corneal Stroma/pathology , Extracellular Matrix Proteins/genetics , Mutation, Missense , Transforming Growth Factor beta/genetics , Adolescent , Aged, 80 and over , Child , Corneal Opacity/diagnosis , DNA Mutational Analysis , Female , Gene Frequency , Heterozygote , Humans , Italy , Male , Mexico , Middle Aged , Pedigree , Polymerase Chain Reaction , Slit Lamp , Young Adult
11.
Br J Ophthalmol ; 101(4): 509-513, 2017 04.
Article in English | MEDLINE | ID: mdl-27402970

ABSTRACT

BACKGROUND/AIMS: To report the clinical, histopathological and genetic features of a variant of lattice corneal dystrophy (LCD) associated with two pathogenic mutations in the transforming growth factor-B-induced (TGFBI) gene. METHODS: Clinical characterisation was performed by slit lamp examination and in vivo confocal microscopic imaging (IVCM). Histopathological characterisation was performed with light microscopic examination of an excised corneal button and a peripheral blood samples were collected for TGFBI screening. RESULTS: A 42-year-old woman presented with progressive photophobia and decreased visual acuity in both eyes. Slit lamp examination demonstrated punctate and linear branching opacities in the mid and posterior corneal stroma, corresponding to hyper-reflective opacities noted on IVCM and amyloid deposition noted on histopathological examination of an excised corneal button. TGFBI screening revealed two previously reported heterozygous missense mutations: c.337G>A (p.(Val113Ile)) in exon 4 and c.1673T>C (p.(Leu558Pro)) in exon 12. Screening of an affected sibling with a similar phenotype revealed that she was also heterozygous for both mutations, while screening of another sibling with punctate but not linear stromal opacities revealed that she was heterozygous for only the p.(Leu558Pro) mutation. CONCLUSIONS: The p.(Val113Ile) mutation results in an alteration of the atypical LCD phenotype associated with the p.(Leu558Pro) mutation. This represents only the second report of the alteration of the phenotype of a TGFBI dystrophy by a second, non-homozygous pathogenic mutation, and thus provides insight into the phenotype-genotype correlation of the TGFBI dystrophies.


Subject(s)
Cornea/pathology , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/genetics , Microscopy, Confocal , Mutation , Photophobia/physiopathology , Adult , Corneal Dystrophies, Hereditary/physiopathology , Disease Progression , Female , Genetic Association Studies , Humans , Pedigree , Phenotype , Photophobia/etiology , Photophobia/genetics
12.
Am J Physiol Cell Physiol ; 311(5): C820-C830, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27581649

ABSTRACT

Congenital hereditary endothelial dystrophy (CHED), Harboyan syndrome (CHED with progressive sensorineural deafness), and potentially a subset of individuals with late-onset Fuchs' endothelial corneal dystrophy are caused by mutations in the SLC4A11 gene that results in corneal endothelial cell abnormalities. Originally classified as a borate transporter, the function of SLC4A11 as a transport protein remains poorly understood. Elucidating the transport function(s) of SLC4A11 is needed to better understand how its loss results in the aforementioned posterior corneal dystrophic disease processes. Quantitative PCR experiments demonstrated that, of the three known human NH2-terminal variants, SLC4A11-C is the major transcript expressed in human corneal endothelium. We studied the expression pattern of the three variants in mammalian HEK-293 cells and demonstrated that the SLC4A11-B and SLC4A11-C variants are plasma membrane proteins, whereas SLC4A11-A is localized intracellularly. SLC4A11-B and SLC4A11-C were shown to be multifunctional ion transporters capable of transporting H+ equivalents in both a Na+-independent and Na+-coupled mode. In both transport modes, SLC4A11-C H+ flux was significantly greater than SLC4A11-B. In the presence of ammonia, SLC4A11-B and SLC4A11-C generated inward currents that were comparable in magnitude. Chimera SLC4A11-C-NH2-terminus-SLC4A11-B experiments demonstrated that the SLC4A11-C NH2-terminus functions as an autoactivating domain, enhancing Na+-independent and Na+-coupled H+ flux without significantly affecting the electrogenic NH3-H(n)+ cotransport mode. All three modes of transport were significantly impaired in the presence of the CHED causing p.R109H (SLC4A11-C numbering) mutation. These complex ion transport properties need to be addressed in the context of corneal endothelial disease processes caused by mutations in SLC4A11.


Subject(s)
Anion Transport Proteins/genetics , Antiporters/genetics , Genetic Variation/genetics , Ion Transport/genetics , Cell Line , Cell Membrane/genetics , Corneal Dystrophies, Hereditary/genetics , Endothelium, Corneal/metabolism , HEK293 Cells , Hearing Loss, Sensorineural/genetics , Humans , Membrane Proteins/genetics , Middle Aged , Mutation/genetics
13.
Invest Ophthalmol Vis Sci ; 57(10): 4136-43, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27537263

ABSTRACT

PURPOSE: To investigate the role of the zinc finger e-box binding homeobox 1 (ZEB1) transcription factor in posterior polymorphous corneal dystrophy 3 by demonstrating its ability to regulate type IV collagen gene transcription via binding to putative E2 box motifs. METHODS: Putative E2 box motifs were identified by in silico analysis within the promoter region of collagen, type IV, alpha3 (COL4A3) and collagen, type IV, alpha4 (COL4A4). To test the ability of ZEB1 to bind to each identified E2 box, electrophoretic mobility shift assays were performed by incubating ZEB1-enriched nuclear extracts with DIG-labeled probes containing one of each of the identified E2 box motifs. Dual-luciferase reporter assays were performed to test the effects of ZEB1 on the luciferase activity of COL4A3 and cadherin 1 (CDH1) promoter constructs, and to determine the effect of a ZEB1 truncating mutation on CDH1 promoter activity. RESULTS: ZEB1 exhibited binding to six of the nine COL4A3 E2 box probes, whereas no binding was observed for either of the two COL4A4 E2 box probes. ZEB1 overexpression resulted in reduced activity of the COL4A3 promoter construct containing all identified E2 box motifs, whereas a truncating ZEB1 mutation led to the loss of ZEB1-dependent repression of the CDH1 promoter. CONCLUSIONS: COL4A3 gene expression is negatively regulated by ZEB1 binding to E2 box motifs in the COL4A3 promoter region. Therefore, the altered expression of type IV collagens, particularly COL4A3, in the corneal endothelium in individuals with PPCD3 is likely due to reduced transcriptional repression in the setting of a single functional ZEB1 allele.


Subject(s)
Autoantigens/genetics , Collagen Type IV/genetics , Corneal Dystrophies, Hereditary/genetics , DNA/genetics , Endothelium, Corneal/metabolism , Gene Expression Regulation , Zinc Finger E-box-Binding Homeobox 1/genetics , Autoantigens/biosynthesis , Cells, Cultured , Collagen Type IV/biosynthesis , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , Electrophoretic Mobility Shift Assay , Endothelium, Corneal/pathology , Epitopes , Humans , Immunoblotting , Promoter Regions, Genetic , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1/biosynthesis , Zinc Fingers
14.
J Ophthalmol ; 2016: 1968493, 2016.
Article in English | MEDLINE | ID: mdl-27382485

ABSTRACT

Purpose. To report the identification of the first de novo UBIAD1 missense mutation in an individual with Schnyder corneal dystrophy (SCD). Methods. A slit lamp examination was performed on a 47-year-old woman without a family history of corneal disorders. The proband's parents, two sisters, and son were also examined and genomic DNA from all six individuals was collected. The exons and exon-intron boundaries of UBIAD1 were screened using Sanger sequencing. Identified mutations were screened for in 200 control chromosomes. In silico analysis predicted the impact of identified mutations on protein function and structure. Results. Slit lamp examination of the proband revealed findings consistent with SCD. Corneas of the family members appeared unaffected. Screening of UBIAD1 in the proband identified a novel heterozygous c.308C>T mutation, predicted to encode the missense amino acid substitution p.(Thr103Ile). This mutation was not identified in any of the family members or in 200 control chromosomes and was predicted to be damaging to normal protein function and structure. Conclusions. We present a novel heterozygous de novo missense mutation in UBIAD1, p.(Thr103Ile), identified in a patient with classic clinical features of SCD. This highlights the value of genetic testing in clinical diagnostic settings, even in the absence of a positive family history.

15.
PLoS One ; 11(6): e0158467, 2016.
Article in English | MEDLINE | ID: mdl-27355326

ABSTRACT

Posterior polymorphous corneal dystrophy 1 (PPCD1) is a genetic disorder that affects corneal endothelial cell function and leads to loss of visual acuity. PPCD1 has been linked to a locus on chromosome 20 in multiple families; however, Sanger sequencing of protein-coding genes in the consensus region failed to identify any causative missense mutations. In this study, custom capture probes were utilized for targeted next-generation sequencing of the linked region in a previously reported family with PPCD1. Variants were detected through two bioinformatics pipelines and filtered according to multiple criteria. Additionally, a high-resolution microarray was used to detect copy number variations. No non-synonymous variants in the protein-coding region of annotated genes were identified. However, 12 single nucleotide variants in 10 genes, and 9 indels in 7 genes met the filtering criteria and were considered candidate variants for PPCD1. Eleven single nucleotide variants were confirmed by Sanger sequencing, including 2 synonymous variants and 9 non-coding variants, in 9 genes. One microdeletion was detected in an intron of OVOL2 by microarray but was subsequently not identified by PCR. Using a comprehensive next-generation sequencing approach, a total of 16 genes containing single nucleotide variants or indels that segregated with the affected phenotype in an affected family previously mapped to the PPCD1 locus were identified. Screening of these candidate genes in other families previously mapped to the PPCD1 locus will likely result in the identification of the genetic basis of PPCD1.


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Polymorphism, Single Nucleotide , Algorithms , Computational Biology , DNA Copy Number Variations , Family Health , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Mutation, Missense , Promoter Regions, Genetic , Thrombomodulin/genetics , Transcription Factors/genetics
16.
PLoS One ; 11(6): e0157418, 2016.
Article in English | MEDLINE | ID: mdl-27309958

ABSTRACT

PURPOSE: To report identification of a COL17A1 mutation in a family with a corneal dystrophy previously mapped to chromosome 10q23-q24. METHODS: Whole-exome sequencing was performed on DNA samples from five affected family members and two unrelated, unaffected individuals. Identified variants were filtered for those that were: located in the linked interval on chromosome 10q23-q24; novel or rare (minor allele frequency ≤0.01); heterozygous; present in all affected individuals and not in controls; and present in genes that encode proteins expressed in human corneal epithelial cells (reads per kilobase per million ≥1). Sanger sequencing of identified variants (SNVs) was performed in additional family members. In silico analysis was used to predict the functional impact of non-synonymous variants. RESULTS: Three SNVs located in two genes were identified that met the filtering criteria: one rare synonymous c.3156C>T variant in the collagen, type XVII, alpha I (COL17A1) gene; and two rare variants, one synonymous and one missense, in the dynamin binding protein (DNMBP) gene. Sanger sequencing of additional family members determined that only the COL17A1 variant segregates with the affected phenotype. In silico analysis predicts that the missense variant in DNMBP would be tolerated. CONCLUSIONS: The corneal dystrophy mapped to chromosome 10q23-q24 is associated with the c.3156C>T variant in COL17A1. As this variant has recently been identified in five other families with early onset recurrent corneal erosions, and has been shown in vitro to introduce a cryptic splice donor site, this dystrophy is likely caused by aberrant splicing of COL17A1 and should be classified as epithelial recurrent erosion dystrophy.


Subject(s)
Alternative Splicing , Autoantigens/genetics , Chromosomes, Human, Pair 10/chemistry , Corneal Dystrophies, Hereditary/genetics , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , Mutation , Non-Fibrillar Collagens/genetics , Aged , Alleles , Autoantigens/metabolism , Case-Control Studies , Chromosome Mapping , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , Cytoskeletal Proteins/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Exome , Female , Gene Expression , Gene Frequency , Genes, Dominant , Genome-Wide Association Study , Heterozygote , Humans , Male , Non-Fibrillar Collagens/metabolism , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Collagen Type XVII
17.
Cell Transplant ; 25(6): 1159-76, 2016.
Article in English | MEDLINE | ID: mdl-26337789

ABSTRACT

The corneal endothelium plays a primary role in maintaining corneal homeostasis and clarity and must be surgically replaced with allogenic donor corneal endothelium in the event of visually significant dysfunction. However, a worldwide shortage of donor corneal tissue has led to a search for alternative sources of transplantable tissue. Cultured human corneal endothelial cells (HCEnC) have been shown to restore corneal clarity in experimental models of corneal endothelial dysfunction in animal models, but characterization of cultured HCEnC remains incomplete. To this end, we utilized next-generation RNA sequencing technology to compare the transcriptomic profile of ex vivo human corneal endothelial cells (evHCEnC) with that of primary HCEnC (pHCEnC) and HCEnC lines and to determine the utility of cultured and immortalized corneal endothelial cells as models of in vivo corneal endothelium. Multidimensional analyses of the transcriptome data sets demonstrated that primary HCEnC have a closer relationship to evHCEnC than do immortalized HCEnC. Subsequent analyses showed that the majority of the genes specifically expressed in HCEnC (not expressed in ex vivo corneal epithelium or fibroblasts) demonstrated a marked variability of expression in cultured cells compared with evHCEnC. In addition, genes associated with either corneal endothelial cell function or corneal endothelial dystrophies were investigated. Significant differences in gene expression and protein levels were observed in the cultured cells compared with evHCEnC for each of the genes tested except for AGBL1 and LOXHD1, which were not detected by RNA-seq or qPCR. Our transcriptomic analysis suggests that at a molecular level pHCEnC most closely resemble evHCEnC and thus represent the most viable cell culture-based therapeutic option for managing corneal endothelial cell dysfunction. Our findings also suggest that investigators should perform an assessment of the entire transcriptome of cultured HCEnC prior to determination of their potential clinical utility for the management of corneal endothelial cell failure.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Corneal/cytology , Gene Expression Profiling/methods , Adolescent , Adult , Aged , Cell Shape , Cells, Cultured , Child , Cluster Analysis , Corneal Dystrophies, Hereditary/genetics , Databases, Genetic , Gene Expression Regulation , Humans , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Sequence Analysis, RNA , Tissue Donors , Young Adult
18.
Ophthalmic Genet ; 37(1): 76-80, 2016.
Article in English | MEDLINE | ID: mdl-24555743

ABSTRACT

BACKGROUND: Hereditary benign intraepithelial dyskeratosis (HBID) is a rare autosomal-dominant disorder of the conjunctiva and oral mucosa first described in and predominantly affecting descendents of Haliwa-Saponi Native Americans. We report a spontaneous case of histopathologically-confirmed HBID affecting an individual not of Native American ancestry. MATERIALS AND METHODS: Report of a case with histopathologic examination of an excised conjunctival specimen as well as molecular and cytogenetic analysis. RESULTS: A Caucasian boy with a history of oral lesions and conjunctival injection from birth developed bilateral corneal opacities at age 5 and underwent penetrating keratoplasty, with recurrence of the corneal opacification shortly after surgery. Examination of a conjunctival biopsy specimen revealed features consistent with HBID. Copy number variant (CNV) analysis revealed a de novo 4q35 duplication that overlapped the duplication previously associated with HBID, although no genes were identified in the common interval. NLRP1 gene sequencing failed to reveal a presumed pathogenic variant. CONCLUSIONS: HBID may develop de novo in individuals who are not of Native American ancestry. The absence of coding regions in a duplicated region of 4q35 common to both the individual that we report and previously associated with HBID raises questions regarding the significance of this CNV in the pathogenesis of HBID.


Subject(s)
Chromosome Duplication/genetics , Chromosomes, Human, Pair 4/genetics , Conjunctival Diseases/diagnosis , Corneal Opacity/diagnosis , Epithelium/abnormalities , Mouth Diseases/diagnosis , Skin Abnormalities/diagnosis , White People , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Child , Conjunctival Diseases/genetics , Corneal Opacity/genetics , DNA Copy Number Variations/genetics , Exome/genetics , Humans , Male , Mouth Diseases/genetics , NLR Proteins , Pedigree , Polymorphism, Single Nucleotide , Skin Abnormalities/genetics
19.
Mol Vis ; 21: 1093-100, 2015.
Article in English | MEDLINE | ID: mdl-26396486

ABSTRACT

PURPOSE: To report the identification of a novel frameshift mutation and copy number variation (CNV) in PIKFYVE in two probands with fleck corneal dystrophy (FCD). METHODS: Slit-lamp examination was performed to identify characteristic features of FCD. After genomic DNA was collected, PCR amplification and automated sequencing of all 41 exons of PIKFYVE was performed. Using genomic DNA, quantitative PCR (qPCR) was performed to detect CNVs within PIKFYVE. RESULTS: In the first FCD proband, numerous panstromal punctate opacities were observed in each of the proband's corneas, consistent with the diagnosis of FCD. Screening of PIKFYVE demonstrated a novel heterozygous frameshift mutation in exon 19, c.3151dupA, which is predicted to encode for a truncated PIKFYVE protein, p.(Asp1052Argfs*18). This variant was identified in an affected sister but not in the proband's unaffected mother or brother or 200 control chromosomes. The second FCD proband presented with bilateral, discrete, punctate, grayish-white stromal opacities. Exonic screening of PIKFYVE revealed no causative variant. However, CNV analysis demonstrated the hemizygous deletion of exons 15 and 16. CONCLUSIONS: We report a novel heterozygous frameshift mutation (c.3151dupA) and a CNV in PIKFYVE, representing the first CNV and the fifth frameshift mutation associated with FCD.


Subject(s)
Base Sequence , Corneal Dystrophies, Hereditary/genetics , DNA Copy Number Variations , Frameshift Mutation , Phosphatidylinositol 3-Kinases/genetics , Sequence Deletion , Adult , Cornea/metabolism , Cornea/pathology , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/pathology , DNA Mutational Analysis , Exons , Female , Gene Expression , Heterozygote , Humans , Molecular Sequence Data
20.
Mol Vis ; 21: 1378-86, 2015.
Article in English | MEDLINE | ID: mdl-26788030

ABSTRACT

PURPOSE: To report potentially pathogenic mutations in the keratin 3 (KRT3) and keratin 12 (KRT12) genes in two individuals with clinically diagnosed Meesmann corneal dystrophy (MECD). METHODS: Slit-lamp examination was performed on the probands and available family members to identify characteristic features of MECD. After informed consent was obtained, saliva samples were obtained as a source of genomic DNA, and screening of KRT3 and KRT12 was performed. Potentially pathogenic variants were screened for in 200 control chromosomes. PolyPhen-2, SIFT, and PANTHER were used to predict the functional impact of identified variants. Short tandem repeat genotyping was performed to confirm paternity. RESULTS: Slit-lamp examination of the first proband demonstrated bilateral, diffusely distributed, clear epithelial microcysts, consistent with MECD. Screening of KRT3 revealed a heterozygous missense variant in exon 1, c.250C>T (p.(Arg84Trp)), which has a minor allele frequency of 0.0076 and was not identified in 200 control chromosomes. In silico analysis with PolyPhen-2 and PANTHER predicted the variant to be damaging to protein function; however, SIFT analysis predicted tolerance of the variant. The second proband demonstrated bilateral, diffusely distributed epithelial opacities that appeared gray-white on direct illumination and translucent on retroillumination. Neither parent demonstrated corneal opacities. Screening of KRT12 revealed a novel heterozygous insertion/deletion variant in exon 6, c.1288_1293delinsAGCCCT (p.(Arg430_Arg431delinsSerPro)). This variant was not present in either of the proband's parents or in 200 control chromosomes and was predicted to be damaging by PolyPhen-2, PANTHER, and SIFT. Haplotype analysis confirmed paternity of the second proband, indicating that the variant arose de novo. CONCLUSIONS: We present a novel KRT12 mutation, representing the first de novo mutation and the first indel in KRT12 associated with MECD. In addition, we report a variant of uncertain significance in KRT3 in an individual with MECD. Although the potential pathogenicity of this variant is unknown, it is the first variant affecting the head domain of K3 to be reported in an individual with MECD and suggests that disease-causing variants associated with MECD may not be restricted to primary sequence alterations of either the helix-initiation or helix-termination motifs of K3 and K12.


Subject(s)
Corneal Dystrophy, Juvenile Epithelial of Meesmann/genetics , Keratin-12/genetics , Keratin-3/genetics , Mutation , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Child , Corneal Dystrophy, Juvenile Epithelial of Meesmann/pathology , DNA Mutational Analysis , Female , Heterozygote , Humans , INDEL Mutation , Keratin-12/chemistry , Keratin-3/chemistry , Male , Middle Aged , Mutation, Missense , Pedigree , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL
...