1
|
Abstract
Keratoconus, a progressive corneal ectasia, is a complex disease with both genetic and environmental risk factors. The exact etiology is not known and is likely variable between individuals. Conditions such as hay fever and allergy are associated with increased risk, while diabetes may be protective. Behaviors such as eye rubbing are also implicated, but direct causality has not been proven. Genetics plays a major role in risk for some individuals, with many large pedigrees showing autosomal inheritance patterns. Several genes have been implicated using linkage and follow-up sequencing in these families. Genome-wide association studies for keratoconus and for quantitative traits such as central corneal thickness have identified several genetic loci that contribute to a cumulative risk for keratoconus, even in people without a family history of the disease. Identification of risk genes for keratoconus is improving our understanding of the biology of this complex disease.
Collapse
Affiliation(s)
- Sionne E M Lucas
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7001, Australia;
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7001, Australia;
| |
Collapse
|
2
|
Litke AM, Samuelson S, Delaney KR, Sauvé Y, Chow RL. Investigating the Pathogenicity of VSX1 Missense Mutations and Their Association With Corneal Disease. Invest Ophthalmol Vis Sci 2019; 59:5824-5835. [PMID: 30535423 DOI: 10.1167/iovs.18-25490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Despite numerous studies associating Visual System Homeobox 1 (VSX1), with posterior polymorphous corneal dystrophy and keratoconus, its role in these diseases is unclear. Here we examine the pathogenicity of VSX1 missense mutations in vitro and in a mouse genetic model. Methods Vsx1 transcriptional repressor activity, protein stability, and subcellular localization activity, was examined using luciferase reporter-based assays, western blotting and immunolabeling, respectively, in transfected human embryonic kidney 293T cells. A genetic model for VSX1 p.P247R was generated to investigate pathogenicity of the mutation, in vivo. A wholemount confocal imaging approach on unfixed intact eyes was developed to examine corneal morphology, curvature, and thickness. Immunolabeling and electroretinography was used to examine retinal phenotype. Results A mutation corresponding to human VSX1 p.P247R led to enhanced transcriptional repressor activity, in vitro. A mouse model for VSX1 p.P247R did not have any observable corneal defect, but did exhibit an abnormal electroretinogram response characterized by a more prominent ON as opposed to OFF panretinal responsiveness. In vitro analysis of additional VSX1 missense mutations showed that they either enhanced repressor activity or did not alter activity. Conclusions Our results indicate that although VSX1 sequence variants can alter transcriptional activity, in the context of a mouse genetic model, at least one of these changes does not lead to corneal abnormalities. While we cannot exclude a role for VSX1 as a risk factor for corneal disease, on its own, it does not appear to play a major causative role.
Collapse
Affiliation(s)
| | - Sarah Samuelson
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Kerry R Delaney
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| | - Yves Sauvé
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Robert L Chow
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
3
|
Loukovitis E, Sfakianakis K, Syrmakesi P, Tsotridou E, Orfanidou M, Bakaloudi DR, Stoila M, Kozei A, Koronis S, Zachariadis Z, Tranos P, Kozeis N, Balidis M, Gatzioufas Z, Fiska A, Anogeianakis G. Genetic Aspects of Keratoconus: A Literature Review Exploring Potential Genetic Contributions and Possible Genetic Relationships with Comorbidities. Ophthalmol Ther 2018; 7:263-292. [PMID: 30191404 PMCID: PMC6258591 DOI: 10.1007/s40123-018-0144-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Indexed: 01/24/2023] Open
Abstract
INTRODUCTION Keratoconus (KC) is a complex, genetically heterogeneous, multifactorial degenerative disorder that is accompanied by corneal ectasia which usually progresses asymmetrically. With an incidence of approximately 1 per 2000 and 2 cases per 100,000 population presenting annually, KC follows an autosomal recessive or dominant pattern of inheritance and is, apparently, associated with genes that interact with environmental, genetic, and/or other factors. This is an important consideration in refractive surgery in the case of familial KC, given the association of KC with other genetic disorders and the imbalance between dizygotic twins. The present review attempts to identify the genetic loci contributing to the different KC clinical presentations and relate them to the common genetically determined comorbidities associated with KC. METHODS The PubMed, MEDLINE, Google Scholar, and GeneCards databases were screened for KC-related articles published in English between January 2006 and November 2017. Keyword combinations of "keratoconus," "risk factor(s)," "genetics," "genes," "genetic association(s)," and "cornea" were used. In total, 217 articles were retrieved and analyzed, with greater weight placed on the more recent literature. Further bibliographic research based on the 217 articles revealed another 124 relevant articles that were included in this review. Using the reviewed literature, an attempt was made to correlate genes and genetic risk factors with KC characteristics and genetically related comorbidities associated with KC based on genome-wide association studies, family-based linkage analysis, and candidate-gene approaches. RESULTS An association matrix between known KC-related genes and KC symptoms and/or clinical signs together with an association matrix between identified KC genes and genetically related KC comorbidities/syndromes were constructed. CONCLUSION Twenty-four genes were identified as potential contributors to KC and 49 KC-related comorbidities/syndromes were found. More than 85% of the known KC-related genes are involved in glaucoma, Down syndrome, connective tissue disorders, endothelial dystrophy, posterior polymorphous corneal dystrophy, and cataract.
Collapse
Affiliation(s)
| | - Konstantinos Sfakianakis
- Division of Surgical Anatomy, Laboratory of Anatomy, Medical School, Democritus University of Thrace, University Campus, Alexandroupolis, Greece
| | - Panagiota Syrmakesi
- AHEPA University Hospital, Thessaloníki, Greece
- Ophthalmica Eye Institute, Thessaloníki, Greece
| | - Eleni Tsotridou
- Ophthalmica Eye Institute, Thessaloníki, Greece
- Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloníki, Greece
| | - Myrsini Orfanidou
- Ophthalmica Eye Institute, Thessaloníki, Greece
- Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloníki, Greece
| | - Dimitra Rafailia Bakaloudi
- Ophthalmica Eye Institute, Thessaloníki, Greece
- Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloníki, Greece
| | - Maria Stoila
- Ophthalmica Eye Institute, Thessaloníki, Greece
- Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloníki, Greece
| | - Athina Kozei
- Ophthalmica Eye Institute, Thessaloníki, Greece
- School of Pharmacology, University of Nicosia, Makedonitissis, Nicosia, Cyprus
| | | | | | | | | | | | - Zisis Gatzioufas
- Department of Ophthalmology, Cornea, Cataract and Refractive Surgery, University Hospital Basel, Basel, Switzerland
| | - Aliki Fiska
- Laboratory of Anatomy, Medical School, Democritus University of Thrace, University Campus, Alexandroupolis, Greece
| | | |
Collapse
|
4
|
Chung DD, Frausto RF, Cervantes AE, Gee KM, Zakharevich M, Hanser EM, Stone EM, Heon E, Aldave AJ. Confirmation of the OVOL2 Promoter Mutation c.-307T>C in Posterior Polymorphous Corneal Dystrophy 1. PLoS One 2017; 12:e0169215. [PMID: 28046031 PMCID: PMC5207508 DOI: 10.1371/journal.pone.0169215] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/13/2016] [Indexed: 01/28/2023] Open
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.
Collapse
Affiliation(s)
- Doug D. Chung
- Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, United States of America
| | - Ricardo F. Frausto
- Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, United States of America
| | - Aleck E. Cervantes
- Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, United States of America
| | - Katherine M. Gee
- Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, United States of America
| | - Marina Zakharevich
- Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, United States of America
| | - Evelyn M. Hanser
- Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, United States of America
| | - Edwin M. Stone
- Department of Ophthalmology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Elise Heon
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Anthony J. Aldave
- Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
5
|
Le DJ, Chung DWD, Frausto RF, Kim MJ, Aldave AJ. Identification of Potentially Pathogenic Variants in the Posterior Polymorphous Corneal Dystrophy 1 Locus. PLoS One 2016; 11:e0158467. [PMID: 27355326 PMCID: PMC4927100 DOI: 10.1371/journal.pone.0158467] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 06/16/2016] [Indexed: 12/13/2022] Open
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.
Collapse
Affiliation(s)
- Derek J. Le
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Duk-Won D. Chung
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Ricardo F. Frausto
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Michelle J. Kim
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Anthony J. Aldave
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
6
|
Hamill CE, Schmedt T, Jurkunas U. Fuchs endothelial cornea dystrophy: a review of the genetics behind disease development. Semin Ophthalmol 2014; 28:281-6. [PMID: 24138036 DOI: 10.3109/08820538.2013.825283] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Fuchs dystrophy represents the most common form of endothelial dystrophy and is a significant cause of visual impairment. The cause of Fuchs dystrophy is a complicated combination of both genetic and environmental factors. Understanding the underlying causes of the disease can potentially lead to new medical treatments preventing loss of vision.
Collapse
Affiliation(s)
- Cecily E Hamill
- Massachusetts Eye and Ear Infirmary , Boston, Massachusetts , USA
| | | | | |
Collapse
|
7
|
Aldave AJ, Ann LB, Frausto RF, Nguyen CK, Yu F, Raber IM. Classification of posterior polymorphous corneal dystrophy as a corneal ectatic disorder following confirmation of associated significant corneal steepening. JAMA Ophthalmol 2014; 131:1583-90. [PMID: 24113819 DOI: 10.1001/jamaophthalmol.2013.5036] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IMPORTANCE The identification of steep corneal curvatures in a significant percentage of patients with posterior polymorphous corneal dystrophy (PPCD) confirms this previously reported association and suggests a role for the ZEB1 protein in keratocyte function. OBJECTIVE To determine whether PPCD is characterized by significant corneal steepening. DESIGN, SETTING, AND PARTICIPANTS Cross-sectional study at university-based and private ophthalmology practices of 38 individuals (27 affected and 11 unaffected) from 23 families with PPCD. EXPOSURE Slitlamp examination and corneal topographic imaging were performed for individuals with PPCD and unaffected family members. Saliva or blood samples were obtained from each individual for DNA isolation and ZEB1 sequencing. Corneal ZEB1 expression was measured using immunohistochemistry. MAIN OUTCOMES AND MEASURES Percentage of individuals affected with PPCD and controls with an average keratometric value greater than 48.0 diopters (D) in each eye; the mean keratometric value averaged for both eyes of individuals with PPCD and controls; and the correlation of ZEB1 mutation with keratometric value. RESULTS ZEB1 coding region mutations were identified in 7 of the 27 affected individuals. Ten of the 38 individuals (26.3%) had average keratometric values greater than 48.0 D OU: 10 of 27 individuals with PPCD (37.0%; 6 of 7 individuals with ZEB1 mutations [85.7%] and 4 of 20 individuals without ZEB1 mutations [20.0%]) and 0 of 11 unaffected individuals (P = .04 for unaffected vs affected individuals; P = .004 for individuals with PPCD with vs without ZEB1 mutation). The mean keratometric value of each eye of affected individuals (48.2 D) was significantly greater than that of each eye of unaffected family members (44.1 D) (P = .03). Affected individuals with ZEB1 mutations demonstrated a mean keratometric value of 53.3 D, which was significantly greater than that of affected individuals without ZEB1 mutations (46.5 D; P = .004). Fluorescence immunohistochemistry demonstrated ZEB1 expression in keratocyte nuclei. CONCLUSIONS AND RELEVANCE Abnormally steep corneal curvatures are identified in 37% of all individuals with PPCD and 86% of affected individuals with PPCD secondary to ZEB1 mutations. ZEB1 is present in keratocyte nuclei, suggesting a role for ZEB1 in keratocyte function. Therefore, ZEB1 may play a role in both corneal stromal and endothelial development and function, and PPCD should be considered both an endothelial dystrophy and an ectatic disorder.
Collapse
Affiliation(s)
- Anthony J Aldave
- Jules Stein Eye Institute, David Geffen School of Medicine at University of California, Los Angeles
| | - Lydia B Ann
- Jules Stein Eye Institute, David Geffen School of Medicine at University of California, Los Angeles
| | - Ricardo F Frausto
- Jules Stein Eye Institute, David Geffen School of Medicine at University of California, Los Angeles
| | - Catherine K Nguyen
- Jules Stein Eye Institute, David Geffen School of Medicine at University of California, Los Angeles
| | - Fei Yu
- Jules Stein Eye Institute, David Geffen School of Medicine at University of California, Los Angeles
| | | |
Collapse
|
8
|
Kim NH, Kim MS. The Clinical Features and Progression of the Disease in Posterior Polymorphous Corneal Dystrophy (PPCD). JOURNAL OF THE KOREAN OPHTHALMOLOGICAL SOCIETY 2014. [DOI: 10.3341/jkos.2014.55.3.368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Na Hyun Kim
- Department of Ophthalmology and Visual Science, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Man Soo Kim
- Department of Ophthalmology and Visual Science, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Abstract
The Cornea Society founded an international committee of 17 corneal experts from the USA, Asia and Europe in 2005. The goal of this group was to develop a new international classification of corneal dystrophies (CD) based on modern clinical, histological and genetic knowledge. Both authors are members of this committee. The elaboration of the classification included the correction of many misinterpretations with regard to the different forms of CD which were published in the past literature. In spite of important results concerning the genetic locus and identification of genes and mutations, corneal dystrophies are typically classified with respect to the level of the cornea that is involved. An accurate and up-to-date template for each form of the 25 CDs was created that included the current clinical, histological and genetic information. To indicate the level of evidence supporting the existence of a given dystrophy, the CDs are divided into four different categories. The new international classification of CDs was published in English, Spanish and German as a 40 page supplement with 64 figures, mostly in color. A more detailed description of genetic mutations is included in the appendix.
Collapse
Affiliation(s)
- W Lisch
- Augenärztliche Privatpraxis Hanau, Kurt-Blaum-Platz 8, 63450, Hanau, Deutschland.
| | | |
Collapse
|
10
|
Schmedt T, Silva MM, Ziaei A, Jurkunas U. Molecular bases of corneal endothelial dystrophies. Exp Eye Res 2011; 95:24-34. [PMID: 21855542 DOI: 10.1016/j.exer.2011.08.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 07/18/2011] [Accepted: 08/03/2011] [Indexed: 01/12/2023]
Abstract
The phrase "corneal endothelial dystrophies" embraces a group of bilateral corneal conditions that are characterized by a non-inflammatory and progressive degradation of corneal endothelium. Corneal endothelial cells exhibit a high pump site density and, along with barrier function, are responsible for maintaining the cornea in its natural state of relative dehydration. Gradual loss of endothelial cells leads to an insufficient water outflow, resulting in corneal edema and loss of vision. Since the pathologic mechanisms remain largely unknown, the only current treatment option is surgical transplantation when vision is severely impaired. In the past decade, important steps have been taken to understand how endothelial degeneration progresses on the molecular level. Studies of affected multigenerational families and sporadic cases identified genes and chromosomal loci, and revealed either Mendelian or complex disorder inheritance patterns. Mutations have been detected in genes that carry important structural, metabolic, cytoprotective, and regulatory functions in corneal endothelium. In addition to genetic predisposition, environmental factors like oxidative stress were found to be involved in the pathogenesis of endotheliopathies. This review summarizes and crosslinks the recent progress on deciphering the molecular bases of corneal endothelial dystrophies.
Collapse
Affiliation(s)
- Thore Schmedt
- Schepens Eye Research Institute, Boston, MA 02114, USA
| | | | | | | |
Collapse
|
11
|
Molecular Genetics of Corneal Disease. Cornea 2011. [DOI: 10.1016/b978-0-323-06387-6.00019-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
12
|
Weisenthal RW, Streeten BW. Descemet's Membrane and Endothelial Dystrophies. Cornea 2011. [DOI: 10.1016/b978-0-323-06387-6.00080-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
13
|
Unusual presentation of presumed posterior polymorphous dystrophy associated with iris heterochromia, band keratopathy, and keratoconus. Cornea 2010; 29:1180-5. [PMID: 20567203 DOI: 10.1097/ico.0b013e3181d007e1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE To report an unusual presentation of posterior polymorphous corneal dystrophy (PPCD) associated with band keratopathy, iridocorneal adhesions, heterochromia, keratoconus, and confocal microscopic findings suggestive of iridocorneal endothelial syndrome. METHODS Confocal microscopy, corneal topography, electroretinography, and genetic analysis were performed in the proband and his siblings. RESULTS A 23-year-old man presented with decreased vision in both eyes over 9 months. Examination revealed bilateral alterations in corneal endothelial mosaic with corneal edema and beaten metal appearance in the right eye and cystoid endothelial opacities in the left eye. Marked heterochromia, band keratopathy, and broad peripheral anterior synechiae were present in both eyes. Topographic features of keratoconus were noted. Electroretinography did not detect abnormal retinal function, as has been described with PPCD associated with VSX1 mutations. Diagnosis of PPCD was postulated on the basis of the examination of 3 of proband's brothers by confocal microscopy. Genetic analysis of 3 known PPCD genes, VSX1, COL8A2, and TCF8, did not detect any mutations. CONCLUSIONS In severe cases, PPCD can resemble iridocorneal endothelial syndromes in both clinical appearance and imaging studies (confocal microscopy). There was a strong genetic phenotypic penetrance in the family, which was essential in the diagnostic decision making. A yet undetermined genotype is contributing to this unusual PPCD phenotype.
Collapse
|
14
|
Abstract
The corneal endothelium maintains the level of hydration in the cornea. Dysfunction of the endothelium results in excess accumulation of water in the corneal stroma, leading to swelling of the stroma and loss of transparency. There are four different corneal endothelial dystrophies that are hereditary, progressive, non-inflammatory disorders involving dysfunction of the corneal endothelium. Each of the endothelial dystrophies is genetically heterogeneous with different modes of transmission and/or different genes involved in each subtype. Genes responsible for disease have been identified for only a subset of corneal endothelial dystrophies. Knowledge of genes involved and their function in the corneal endothelium can aid understanding the pathogenesis of the disorder as well as reveal pathways that are important for normal functioning of the endothelium.
Collapse
|
15
|
Mutational screening of VSX1 in keratoconus patients from the European population. Eye (Lond) 2009; 24:1085-92. [DOI: 10.1038/eye.2009.217] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
16
|
Weiss JS, Møller HU, Lisch W, Kinoshita S, Aldave AJ, Belin MW, Kivelä T, Busin M, Munier FL, Seitz B, Sutphin J, Bredrup C, Mannis MJ, Rapuano CJ, Van Rij G, Kim EK, Klintworth GK. The IC3D classification of the corneal dystrophies. Cornea 2008; 27 Suppl 2:S1-83. [PMID: 19337156 PMCID: PMC2866169 DOI: 10.1097/ico.0b013e31817780fb] [Citation(s) in RCA: 197] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The recent availability of genetic analyses has demonstrated the shortcomings of the current phenotypic method of corneal dystrophy classification. Abnormalities in different genes can cause a single phenotype, whereas different defects in a single gene can cause different phenotypes. Some disorders termed corneal dystrophies do not appear to have a genetic basis. PURPOSE The purpose of this study was to develop a new classification system for corneal dystrophies, integrating up-to-date information on phenotypic description, pathologic examination, and genetic analysis. METHODS The International Committee for Classification of Corneal Dystrophies (IC3D) was created to devise a current and accurate nomenclature. RESULTS This anatomic classification continues to organize dystrophies according to the level chiefly affected. Each dystrophy has a template summarizing genetic, clinical, and pathologic information. A category number from 1 through 4 is assigned, reflecting the level of evidence supporting the existence of a given dystrophy. The most defined dystrophies belong to category 1 (a well-defined corneal dystrophy in which a gene has been mapped and identified and specific mutations are known) and the least defined belong to category 4 (a suspected dystrophy where the clinical and genetic evidence is not yet convincing). The nomenclature may be updated over time as new information regarding the dystrophies becomes available. CONCLUSIONS The IC3D Classification of Corneal Dystrophies is a new classification system that incorporates many aspects of the traditional definitions of corneal dystrophies with new genetic, clinical, and pathologic information. Standardized templates provide key information that includes a level of evidence for there being a corneal dystrophy. The system is user-friendly and upgradeable and can be retrieved on the website www.corneasociety.org/ic3d.
Collapse
Affiliation(s)
- Jayne S Weiss
- Department of Ophthalmology, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Mok JW, Baek SJ, Joo CK. VSX1 gene variants are associated with keratoconus in unrelated Korean patients. J Hum Genet 2008; 53:842-849. [PMID: 18626569 DOI: 10.1007/s10038-008-0319-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 06/23/2008] [Indexed: 10/21/2022]
Abstract
Keratoconus is a bilateral ectatic disorder characterized by the central thinning of corneal tissue leading to visual impairment. To investigate the possibility of visual system homeobox 1 (VSXI) as a candidate susceptibility gene for Korean patients with keratoconus, we performed a mutation screening of the VSXI gene in 249 unrelated patients with keratoconus and 208 control subjects without the ocular disorder. We found two heterozygous novel missense mutations in exon 2: N151S and G160V. The G160V mutation was identified in 13 keratoconus patients (5.3%), and the N151S mutation was found in only one keratoconus patient (0.4%). We also detected three synonymous polymorphisms and four intragenic polymorphisms. The IVS1-11*a allele was associated with a significantly increased risk of keratoconus in Korean patients [3.6 vs. 0.5%, p = 0.001, odds ratio (OR) = 7.76, 95% confidence interval (CI) 1.989-30.241). Other polymorphisms did not show an association with keratoconus risk. Our data is the first reported VSX1 mutation screening in Korean keratoconus patients. We detected two novel missense mutations and one intragenic polymorphism in the VSX1 gene, which show a strong statistical association with unrelated keratoconus patients. Consequently, our study suggests that VSX1 gene variants seem to be significant genetic variants for keratoconus predisposition in unrelated Korean patients.
Collapse
Affiliation(s)
- Jee-Won Mok
- Laboratory of Ophthalmology and Visual Science, Department of Ophthalmology and Visual Science, Catholic Research Institutes of Medical Science, Catholic University Medical College, The Catholic University of Korea, 505 Banpo-dong, Seocho-ku, Seoul, 137-701, South Korea.,Korea Eye Tissue and Gene Bank, St. Mary's Hospital, Seoul, South Korea
| | - Sun-Jin Baek
- Laboratory of Ophthalmology and Visual Science, Department of Ophthalmology and Visual Science, Catholic Research Institutes of Medical Science, Catholic University Medical College, The Catholic University of Korea, 505 Banpo-dong, Seocho-ku, Seoul, 137-701, South Korea.,Korea Eye Tissue and Gene Bank, St. Mary's Hospital, Seoul, South Korea
| | - Choun-Ki Joo
- Laboratory of Ophthalmology and Visual Science, Department of Ophthalmology and Visual Science, Catholic Research Institutes of Medical Science, Catholic University Medical College, The Catholic University of Korea, 505 Banpo-dong, Seocho-ku, Seoul, 137-701, South Korea. .,Korea Eye Tissue and Gene Bank, St. Mary's Hospital, Seoul, South Korea. .,Department of Ophthalmology, St. Mary's Hospital, Seoul, South Korea.
| |
Collapse
|
18
|
Liu Y, Peng X, Tan J, Darling DS, Kaplan HJ, Dean DC. Zeb1 mutant mice as a model of posterior corneal dystrophy. Invest Ophthalmol Vis Sci 2008; 49:1843-9. [PMID: 18436818 DOI: 10.1167/iovs.07-0789] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The zinc finger transcription factor Zeb1 binds to E-box-like sequences and is important for maintaining repression of epithelial specification genes in vivo. Overexpression of Zeb1 in cancer triggers epithelial-mesenchymal transition, which facilitates metastasis. The mutation of ZEB1 in humans is linked to posterior polymorphous corneal dystrophy (PPCD), in which an epithelial transition of the corneal endothelium is associated with abnormal endothelial proliferation. The purpose of this study is to determine whether Zeb1 null or heterozygous mice may provide an animal model for PPCD. METHODS Corneal morphology, protein and mRNA expression, and cell proliferation were compared in wild-type and Zeb1 gene knockout mice by immunostaining, real-time PCR, and BrdU incorporation. mRNA expression in isolated embryo fibroblasts derived from wild-type, Zeb1 heterozygous, and null mice was analyzed by real-time PCR RESULTS: Zeb1 null mice late in gestation show ectopic expression of epithelial genes in the corneal endothelium and keratocytes, including the basement membrane component COL4A3, which is ectopically expressed by the corneal endothelium in PPCD. These embryos also show abnormal corneal endothelial and keratocyte proliferation, corneal thickening, and corneolenticular and iridocorneal adhesions. Adult Zeb1 heterozygous mice exhibit these same corneal defects. The ectopic expression of epithelial genes extended to embryonic fibroblasts derived from Zeb1 heterozygous and null mice, suggesting that Zeb1 may have a more general role in the suppression of an epithelial phenotype. CONCLUSIONS The authors conclude that Zeb1 heterozygous and null mice show features of PPCD and thus should provide an animal model for genetic dissection of pathways contributing to the disease.
Collapse
Affiliation(s)
- Yongqing Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, 301 E. Muhammad Ali Boulevard, Louisville, KY 40202, USA
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
PURPOSE Three mutations, L159M, R166W, and H244R, in the VSX1 gene have been recently reported to be associated with keratoconus by direct sequencing in familial panels. In an attempt to confirm this observation, we surveyed the same mutations of the VSX1 gene for a white sporadic keratoconus case-control panel and a larger familial panel to test its association with keratoconus. METHODS A case-control panel, with 77 keratoconus patients and 71 healthy controls, and a keratoconus familial panel, with 444 individuals from 75 families, were surveyed. DNA from each individual was tested for the previously reported mutations by ABI allelic discrimination technology (L159M and R166W) and restriction fragment length polymorphism assay (H244R). RESULTS We observed no mutations of R166W and H244R and 1 heterozygous mutation of L159M in a healthy individual in the case-control panel. For the familial panel, we observed no polymorphism of R166W; 3 heterozygous for H244R, with 2 affected and 1 unaffected; and 5 heterozygous for L159M, with 3 affected and 2 unaffected. CONCLUSIONS We cannot confirm the previously reported association of the polymorphism in the VSX1 gene with keratoconus. In our case-control sample panel and the larger familial sample panel, we did not observe the reported polymorphism of the VSX1 gene, and the distribution of these 3 polymorphisms was not significant enough to support a pathogenetic role in keratoconus.
Collapse
|
20
|
Aldave AJ, Yellore VS, Yu F, Bourla N, Sonmez B, Salem AK, Rayner SA, Sampat KM, Krafchak CM, Richards JE. Posterior polymorphous corneal dystrophy is associated with TCF8 gene mutations and abdominal hernia. Am J Med Genet A 2008; 143A:2549-56. [PMID: 17935237 DOI: 10.1002/ajmg.a.31978] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Mutations in the two-handed zinc-finger homeodomain transcription factor gene (TCF8) have been associated with posterior polymorphous corneal dystrophy (PPCD) and extraocular developmental abnormalities. We performed screening of TCF8 in 32 affected, unrelated probands, affected and unaffected family members of probands identified with a TCF8 mutation, and in 100 control individuals. Eight different pathogenic mutations were identified in eight probands: four frameshift (c.953_954insA, c.1506dupA, c.1592delA, and c.3012_3013delAG); three nonsense (Gln12X, Gln214X, Arg325X); and one missense (Met1Arg). Screening of TCF8 in affected and unaffected family members in six families demonstrated that each identified mutation segregated with the disease phenotype in each family; two probands did not have additional family members available for analysis. None of the eight TCF8 mutations was identified in 200 control chromosomes. The prevalence of hernias of the abdominal region in affected individuals with PPCD associated with TCF8 mutations was significantly higher than the prevalence in both individuals with PPCD not associated with a TCF8 mutation and in unaffected individuals. Therefore, PPCD is associated with TCF8 mutations in one quarter of affected families in this study, or about one third of all PPCD families that have been screened thus far. In these families, the presence of apparently causative TCF8 mutations is associated with abdominal and inguinal hernias.
Collapse
Affiliation(s)
- Anthony J Aldave
- The Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Yellore VS, Papp JC, Sobel E, Khan MA, Rayner SA, Farber DB, Aldave AJ. Replication and refinement of linkage of posterior polymorphous corneal dystrophy to the posterior polymorphous corneal dystrophy 1 locus on chromosome 20. Genet Med 2007; 9:228-34. [PMID: 17438387 DOI: 10.1097/gim.0b013e31803c4dc2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
PURPOSE The study purpose was to identify the genetic basis of posterior polymorphous corneal dystrophy, an autosomal dominant disorder of the corneal endothelium that is associated with the development of corneal edema, necessitating corneal transplantation for visual rehabilitation. Glaucoma also develops in up to 40% of patients with posterior polymorphous corneal dystrophy. METHODS Linkage analysis, using microsatellite markers previously used to demonstrate linkage of posterior polymorphous corneal dystrophy to the chromosome 20 candidate region known as posterior polymorphous corneal dystrophy 1, was performed in 29 members of a family with posterior polymorphous corneal dystrophy. Thirty-four microsatellite markers were used to refine the posterior polymorphous corneal dystrophy 1 interval. TCF8, located on chromosome 10, was screened in an affected family member to exclude posterior polymorphous corneal dystrophy 3. RESULTS Significant evidence of linkage to the posterior polymorphous corneal dystrophy 1 interval was obtained with both single-point and multipoint analyses. The largest single-point log odds ratio score obtained was 4.38 (theta=0) at marker D20S471; within 4.7 Mbp (7.2 cM) of D20S471 eight markers provided single-point log odds ratio scores of greater than 3.00 and three markers provided single-point log odds ratio scores greater than 4.00. The largest multipoint log odds ratio score obtained was 4.83, found across the adjacent markers D20S844, D20S191, D20S484, and D20S111. The support interval for posterior polymorphous corneal dystrophy 1 in the family we report is approximately 13.5 Mbp (10 cM) long and lies between the markers D20S182 and D20S195. Eleven markers have multipoint log odds ratio scores greater than 4.0 within this region. No coding region mutations were identified in TCF8 in an affected member of the family, effectively excluding posterior polymorphous corneal dystrophy 3. CONCLUSIONS The originally described 19.8 cM posterior polymorphous corneal dystrophy 1 candidate disease interval has been refined to a 10 cM interval between markers D20S182 and D20S195. A portion of this refined interval overlaps a more recently reported posterior polymorphous corneal dystrophy 1 interval, with only 20 known and predicted genes mapped to the 2.4 cM common interval.
Collapse
Affiliation(s)
- Vivek S Yellore
- Cornea Service, The Jules Stein Eye Institute, Department of Human Genetics, and Retinal Biochemistry Laboratory, David Geffen School of Medicine at University of California Los Angeles, California 90095, USA
| | | | | | | | | | | | | |
Collapse
|