Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Publication year range
1.
Article in English | MEDLINE | ID: mdl-38599222

ABSTRACT

Glaucoma is a clinically heterogeneous disease and the world's leading cause of irreversible blindness. Therapeutic intervention can prevent blindness but relies on early diagnosis, and current clinical risk factors are limited in their ability to predict who will develop sight-threatening glaucoma. The high heritability of glaucoma makes it an ideal substrate for genetic risk prediction, with the bulk of risk being polygenic in nature. Here, we summarize the foundations of glaucoma genetic risk, the development of polygenic risk prediction instruments, and emerging opportunities for genetic risk stratification. Although challenges remain, genetic risk stratification will significantly improve glaucoma screening and management.

2.
BMC Ophthalmol ; 23(1): 431, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37875865

ABSTRACT

BACKGROUND: Glaucoma and age-related macular degeneration (AMD) account for a substantial portion of global blindness. Both conditions are highly heritable, with recognised monogenic and polygenic inheritance patterns. Current screening guidelines lack decisive recommendations. Polygenic risk scores (PRS) allow for cost-effective broad population risk stratification for these conditions. The predictive potential of PRS could facilitate earlier diagnosis and treatment, and prevent unnecessary vision loss. METHODS: The Genetic Risk Assessment of Degenerative Eye disease (GRADE) study is a prospective study designed to generate high-quality evidence about the feasibility of PRS to stratify individuals from the general population, enabling identification of those at highest risk of developing glaucoma or AMD. The targeted recruitment is 1000 individuals aged over 50 years, from which blood or saliva samples will be used for genotyping and an individual PRS for glaucoma and AMD will be derived. Individuals with PRS values in the bottom decile (n = 100), top decile (n = 100) and middle 80% (n = 100) for both glaucoma and AMD will undergo a detailed eye examination for glaucoma and/or AMD. DISCUSSION: The primary objective will be to compare the prevalence of glaucoma and AMD cases between low, intermediate, and high PRS risk groups. We expect to find a higher prevalence of both diseases in the high PRS risk group, as compared to the middle and low risk groups. This prospective study will assess the clinical validity of a PRS for glaucoma and AMD in the general Australian population. Positive findings will support the implementation of PRS into clinical practice.


Subject(s)
Glaucoma , Macular Degeneration , Humans , Middle Aged , Prospective Studies , Multifactorial Inheritance , Australia , Glaucoma/diagnosis , Glaucoma/genetics , Glaucoma/epidemiology , Risk Factors , Risk Assessment , Macular Degeneration/diagnosis , Macular Degeneration/genetics , Macular Degeneration/epidemiology
3.
Hum Mutat ; 43(12): 2170-2186, 2022 12.
Article in English | MEDLINE | ID: mdl-36217948

ABSTRACT

The standardization of variant curation criteria is essential for accurate interpretation of genetic results and clinical care of patients. The variant curation guidelines developed by the American College of Medical Genetics and Genomics (ACMG) and the Association for Molecular Pathology (AMP) in 2015 are widely used but are not gene specific. To address this issue, the Clinical Genome Resource (ClinGen) Variant Curation Expert Panels (VCEP) have been tasked with developing gene-specific variant curation guidelines. The Glaucoma VCEP was created to develop rule specifications for genes associated with primary glaucoma, including myocilin (MYOC), the most common cause of Mendelian glaucoma. Of the 28 ACMG/AMP criteria, the Glaucoma VCEP adapted 15 rules to MYOC and determined 13 rules not applicable. Key specifications included determining minor allele frequency thresholds, developing an approach to counting probands and segregations, and reviewing functional assays. The rules were piloted on 81 variants and led to a change in classification in 40% of those that were classified in ClinVar, with functional evidence influencing the classification of 18 variants. The standardized variant curation guidelines for MYOC provide a framework for the consistent application of the rules between laboratories, to improve MYOC genetic testing in the management of glaucoma.


Subject(s)
Genome, Human , Glaucoma , Humans , Genetic Testing/methods , Genetic Variation , Glaucoma/diagnosis , Glaucoma/genetics , Pathology, Molecular , United States
4.
Am J Hum Genet ; 104(4): 578-595, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30951675

ABSTRACT

The evidence base supporting genetic and genomic sequence-variant interpretations is continuously evolving. An inherent consequence is that a variant's clinical significance might be reinterpreted over time as new evidence emerges regarding its pathogenicity or lack thereof. This raises ethical, legal, and financial issues as to whether there is a responsibility to recontact research participants to provide updates on reinterpretations of variants after the initial analysis. There has been discussion concerning the extent of this obligation in the context of both research and clinical care. Although clinical recommendations have begun to emerge, guidance is lacking on the responsibilities of researchers to inform participants of reinterpreted results. To respond, an American Society of Human Genetics (ASHG) workgroup developed this position statement, which was approved by the ASHG Board in November 2018. The workgroup included representatives from the National Society of Genetic Counselors, the Canadian College of Medical Genetics, and the Canadian Association of Genetic Counsellors. The final statement includes twelve position statements that were endorsed or supported by the following organizations: Genetic Alliance, European Society of Human Genetics, Canadian Association of Genetic Counsellors, American Association of Anthropological Genetics, Executive Committee of the American Association of Physical Anthropologists, Canadian College of Medical Genetics, Human Genetics Society of Australasia, and National Society of Genetic Counselors.


Subject(s)
Duty to Recontact , Duty to Warn/legislation & jurisprudence , Genetic Testing/standards , Genetics, Medical/standards , Genomics/standards , Australia , Canada , Ethics, Research , Europe , Genetics, Medical/education , Genetics, Medical/ethics , Humans , Liability, Legal , Research Subjects , Societies, Medical , United States
5.
Mol Vis ; 28: 257-268, 2022.
Article in English | MEDLINE | ID: mdl-36284667

ABSTRACT

Purpose: ADAMTSL4-associated ectopia lentis is a rare autosomal recessive condition that is primarily associated with crystalline lens displacement. However, the prevalence of other ocular and systemic manifestations of this condition is poorly understood. In this study, we summarize the ocular and systemic phenotypic spectrum of this condition. Methods: A cross-sectional case study series of four individuals with biallelic pathogenic or likely pathogenic ADAMTSL4 variants was performed alongside a literature review of individuals with ADAMTSL4-associated ectopia lentis on September 29, 2021. Ocular and systemic findings, complications, and genetic findings of all four individuals were collected and summarized. Results: The phenotypic spectrum across 91 individuals sourced from literature and four individuals from this case study series was highly variable. The main ocular phenotypes included ectopia lentis (95/95, 100%), ectopia lentis et pupillae (18/95, 19%), iris transillumination (13/95, 14%), iridodonesis (12/95, 13%), persistent pupillary membrane (12/95, 13%), and early-onset cataract or lens opacities (12/95, 13%). Anterior segment features other than ectopia lentis appeared to be exclusively associated with biallelic loss of function variants (p<0.001). Pupillary block glaucoma had a prevalence of 1%. Post-lensectomy complications included retinal detachment (6/41, 15%), elevated intraocular pressure (4/41, 10%), and aphakic glaucoma (1/41, 2%). Most individuals were not reported to have had systemic features (69/95, 73%). Conclusions: The clinical phenotype of ADAMTSL4-associated ectopia lentis was summarized and expanded. Clinicians should be aware of the varied ocular phenotype and the risks of retinal detachment, ocular hypertension, and glaucoma in the diagnosis and management of this condition.


Subject(s)
Ectopia Lentis , Glaucoma , Retinal Detachment , Humans , Ectopia Lentis/complications , Ectopia Lentis/genetics , Ectopia Lentis/diagnosis , Pedigree , Cross-Sectional Studies , ADAMTS Proteins/genetics , Phenotype , Glaucoma/complications , Glaucoma/genetics
6.
Hum Mutat ; 42(7): 818-826, 2021 07.
Article in English | MEDLINE | ID: mdl-33973672

ABSTRACT

Gelsolin (GSN) variants have been implicated in amyloidosis of the Finnish type. This case series reports a novel GSN:c.1477T>C,p.(Trp493Arg) variant in a family with ocular and systemic features consistent with Finnish Amyloidosis. Exome sequencing performed on affected individuals from two families manifesting cutis laxa and polymorphic corneal stromal opacities demonstrated the classic GSN:c.654G>A,p.Asp214Asn variant in single affected individual from one family, and a previously undocumented GSN:c.1477T>C variant in three affected first-degree relatives from a separate family. Immunohistochemical studies on corneal tissue from a proband with the c.1477T>C variant identified gelsolin protein within histologically defined corneal amyloid deposits. This study reports a novel association between the predicted pathogenic GSN:c.1477T>C variant and amyloidosis of the Finnish type, and is the first to provide functional evidence of a pathological GSN variant at a locus distant to the critical G2 calcium-binding region, resulting in the phenotype of amyloidosis of the Finnish type.


Subject(s)
Amyloidosis , Corneal Dystrophies, Hereditary , Amyloidosis/genetics , Calcium/metabolism , Corneal Dystrophies, Hereditary/genetics , Finland , Gelsolin/genetics , Gelsolin/metabolism , Genetic Variation , Humans
7.
Ophthalmology ; 128(11): 1549-1560, 2021 11.
Article in English | MEDLINE | ID: mdl-33892047

ABSTRACT

PURPOSE: To report the relative frequencies of childhood and early onset glaucoma subtypes and their genetic findings in a large single cohort. DESIGN: Retrospective clinical and molecular study. PARTICIPANTS: All individuals with childhood glaucoma (diagnosed 0 to <18 years) and early onset glaucoma (diagnosed 18 to <40 years) referred to a national disease registry. METHODS: We retrospectively reviewed the referrals of all individuals with glaucoma diagnosed at <40 years of age recruited to the Australian and New Zealand Registry of Advanced Glaucoma (ANZRAG). Subtypes of glaucoma were determined using the Childhood Glaucoma Research Network (CGRN) classification system. DNA extracted from blood or saliva samples underwent sequencing of genes associated with glaucoma. MAIN OUTCOME MEASURES: The phenotype and genotype distribution of glaucoma diagnosed at <40 years of age. RESULTS: A total of 290 individuals (533 eyes) with childhood glaucoma and 370 individuals (686 eyes) with early onset glaucoma were referred to the ANZRAG. Primary glaucoma was the most prevalent condition in both cohorts. In the childhood cohort, 57.6% of individuals (167/290, 303 eyes) had primary congenital glaucoma (PCG), and 19.3% (56/290, 109 eyes) had juvenile open-angle glaucoma. Juvenile open-angle glaucoma constituted 73.2% of the early onset glaucoma cohort (271/370, 513 eyes). Genetic testing in probands resulted in a diagnostic yield of 24.7% (125/506) and a reclassification of glaucoma subtype in 10.4% of probands (13/125). The highest molecular diagnostic rate was achieved in probands with glaucoma associated with nonacquired ocular anomalies (56.5%). Biallelic variants in CYP1B1 (n = 29, 23.2%) and heterozygous variants in MYOC (n = 24, 19.2%) and FOXC1 (n = 21, 16.8%) were most commonly reported among probands with a molecular diagnosis. Biallelic CYP1B1 variants were reported in twice as many female individuals as male individuals with PCG (66.7% vs. 33.3%, P = 0.02). CONCLUSIONS: We report on the largest cohort of individuals with childhood and early onset glaucoma from Australasia using the CGRN classification. Primary glaucoma was most prevalent. Genetic diagnoses ascertained in 24.7% of probands supported clinical diagnoses and genetic counseling. International collaborative efforts are required to identify further genes because the majority of individuals still lack a clear molecular diagnosis.


Subject(s)
Eye Proteins/genetics , Genetic Profile , Glaucoma/classification , Intraocular Pressure/physiology , Mutation , Registries , Adolescent , Australia/epidemiology , Child , Child, Preschool , Eye Proteins/metabolism , Female , Genetic Testing , Genotype , Glaucoma/epidemiology , Glaucoma/genetics , Humans , Infant , Infant, Newborn , Male , New Zealand/epidemiology , Pedigree , Phenotype , Retrospective Studies
8.
Am J Med Genet A ; 185(2): 434-439, 2021 02.
Article in English | MEDLINE | ID: mdl-33231930

ABSTRACT

Axenfeld-Rieger syndrome is a genetic condition characterized by ocular and systemic features and is most commonly caused by variants in the FOXC1 or PITX2 genes. Facial dysmorphism is part of the syndrome but the differences between both genes have never been systematically assessed. Here, 11 facial traits commonly reported in Axenfeld-Rieger syndrome were assessed by five clinical geneticists blinded to the molecular diagnosis. Individuals were drawn from the Australian and New Zealand Registry of Advanced Glaucoma in Australia or recruited through the Genetic and Ophthalmology Unit of l'Azienda Socio-Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda in Italy. Thirty-four individuals from 18 families were included. FOXC1 variants were present in 64.7% of individuals and PITX2 variants in 35.3% of individuals. A thin upper lip (55.9%) and a prominent forehead (41.2%) were common facial features shared between both genes. Hypertelorism/telecanthus (81.8% vs 25.0%, p = 0.002) and low-set ears (31.8% vs 0.0%, p = 0.036) were significantly more prevalent in individuals with FOXC1 variants compared with PITX2 variants. These findings may assist clinicians in reaching correct clinical and molecular diagnoses, and providing appropriate genetic counseling.


Subject(s)
Abnormalities, Multiple/genetics , Anterior Eye Segment/abnormalities , Craniofacial Abnormalities/genetics , Eye Abnormalities/genetics , Eye Diseases, Hereditary/genetics , Forkhead Transcription Factors/genetics , Homeodomain Proteins/genetics , Muscular Atrophy/genetics , Transcription Factors/genetics , Abnormalities, Multiple/epidemiology , Abnormalities, Multiple/pathology , Adolescent , Adult , Aged , Anterior Eye Segment/pathology , Australia/epidemiology , Child , Child, Preschool , Craniofacial Abnormalities/epidemiology , Craniofacial Abnormalities/pathology , Eye Abnormalities/epidemiology , Eye Abnormalities/pathology , Eye Diseases, Hereditary/epidemiology , Eye Diseases, Hereditary/pathology , Female , Genetic Predisposition to Disease , Genotype , Humans , Infant , Italy/epidemiology , Male , Middle Aged , Muscular Atrophy/epidemiology , Muscular Atrophy/pathology , Mutation/genetics , Pedigree , Phenotype , Young Adult , Homeobox Protein PITX2
9.
Ophthalmology ; 127(7): 901-907, 2020 07.
Article in English | MEDLINE | ID: mdl-32081492

ABSTRACT

PURPOSE: To examine the combined effects of common genetic variants associated with intraocular pressure (IOP) on primary open-angle glaucoma (POAG) phenotype using a polygenic risk score (PRS) stratification. DESIGN: Cross-sectional study. PARTICIPANTS: For the primary analysis, we examined the glaucoma phenotype of 2154 POAG patients enrolled in the Australian and New Zealand Registry of Advanced Glaucoma, including patients recruited from the United Kingdom. For replication, we examined an independent cohort of 624 early POAG patients. METHODS: Using IOP genome-wide association study summary statistics, we developed a PRS derived solely from IOP-associated variants and stratified POAG patients into 3 risk tiers. The lowest and highest quintiles of the score were set as the low- and high-risk groups, respectively, and the other quintiles were set as the intermediate risk group. MAIN OUTCOME MEASURES: Clinical glaucoma phenotype including maximum recorded IOP, age at diagnosis, number of family members affected by glaucoma, cup-to-disc ratio, visual field mean deviation, and treatment intensity. RESULTS: A dose-response relationship was found between the IOP PRS and the maximum recorded IOP, with the high genetic risk group having a higher maximum IOP by 1.7 mmHg (standard deviation [SD], 0.62 mmHg) than the low genetic risk group (P = 0.006). Compared with the low genetic risk group, the high genetic risk group had a younger age of diagnosis by 3.7 years (SD, 1.0 years; P < 0.001), more family members affected by 0.46 members (SD, 0.11 members; P < 0.001), and higher rates of incisional surgery (odds ratio, 1.5; 95% confidence interval, 1.1-2.0; P = 0.007). No statistically significant difference was found in mean deviation. We further replicated the maximum IOP, number of family members affected by glaucoma, and treatment intensity (number of medications) results in the early POAG cohort (P ≤ 0.01). CONCLUSIONS: The IOP PRS was correlated positively with maximum IOP, disease severity, need for surgery, and number of affected family members. Genes acting via IOP-mediated pathways, when considered in aggregate, have clinically important and reproducible implications for glaucoma patients and their close family members.


Subject(s)
Genome-Wide Association Study/methods , Glaucoma, Open-Angle/physiopathology , Intraocular Pressure/physiology , Visual Acuity , Cross-Sectional Studies , Female , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/therapy , Humans , Male , Middle Aged , Phenotype , Risk Factors , Visual Fields/physiology
10.
Ophthalmology ; 127(6): 758-766, 2020 06.
Article in English | MEDLINE | ID: mdl-32085876

ABSTRACT

PURPOSE: Developmental abnormalities of the ocular anterior segment in some cases can lead to ocular hypertension and glaucoma. CPAMD8 is a gene of unknown function recently associated with ocular anterior segment dysgenesis, myopia, and ectopia lentis. We sought to assess the contribution of biallelic CPAMD8 variants to childhood and juvenile open-angle glaucoma. DESIGN: Retrospective, multicenter case series. PARTICIPANTS: A total of 268 probands and their relatives with a diagnosis of childhood or juvenile open-angle glaucoma. PURPOSE: Developmental abnormalities of the ocular anterior segment in some cases can lead to ocular hypertension and glaucoma. CPAMD8 is a gene of unknown function recently associated with ocular anterior segment dysgenesis, myopia, and ectopia lentis. We sought to assess the contribution of biallelic CPAMD8 variants to childhood and juvenile open-angle glaucoma. METHODS: Patients underwent a comprehensive ophthalmic assessment, with DNA from patients and their relatives subjected to genome, exome, or capillary sequencing. CPAMD8 RNA expression analysis was performed on tissues dissected from cadaveric human eyes. MAIN OUTCOME MEASURES: Diagnostic yield within a cohort of childhood and juvenile open-angle glaucoma, prevalence and risk of ophthalmic phenotypes, and relative expression of CPAMD8 in the human eye. RESULTS: We identified rare (allele frequency < 4×10-5) biallelic CPAMD8 variants in 5.7% (5/88) of probands with childhood glaucoma and 2.1% (2/96) of probands with juvenile open-angle glaucoma. When including family members, we identified 11 individuals with biallelic variants in CPAMD8 from 7 unrelated families. Nine of these individuals were diagnosed with glaucoma (9/11, 81.8%), with a mean age at diagnosis of 9.22±14.89 years, and all individuals with glaucoma required 1 or more incisional procedures to control high intraocular pressure. Iris abnormalities were observed in 9 of 11 individuals, cataract was observed in 8 of 11 individuals (72.7%), and retinal detachment was observed in 3 of 11 individuals (27.3%). CPAMD8 expression was highest in neural crest-derived tissues of the adult anterior segment, suggesting that CPAMD8 variation may cause malformation or obstruction of key drainage structures. CONCLUSIONS: Biallelic CPAMD8 variation was associated with a highly heterogeneous phenotype and in our cohorts was the second most common inherited cause of childhood glaucoma after CYP1B1 and juvenile open-angle glaucoma after MYOC. CPAMD8 sequencing should be considered in the investigation of both childhood and juvenile open-angle glaucoma, particularly when associated with iris abnormalities, cataract, or retinal detachment.


Subject(s)
Anterior Eye Segment/abnormalities , Complement C3/genetics , Eye Abnormalities/genetics , Glaucoma, Open-Angle/genetics , Polymorphism, Single Nucleotide , Trypsin Inhibitor, Kazal Pancreatic/genetics , alpha-Macroglobulins/genetics , Adolescent , Adult , Child , Child, Preschool , Exome/genetics , Female , Gene Frequency , Humans , Hydrophthalmos/genetics , Infant , Infant, Newborn , Male , Pedigree , Phenotype , RNA/genetics , Retrospective Studies , Sequence Analysis, DNA , Young Adult
11.
Clin Genet ; 97(5): 764-769, 2020 05.
Article in English | MEDLINE | ID: mdl-32052405

ABSTRACT

Nanophthalmos and posterior microphthalmos are ocular abnormalities in which both eyes are abnormally small, and typically associated with extreme hyperopia. We recruited 40 individuals from 13 kindreds with nanophthalmos or posterior microphthalmos, with 12 probands subjected to exome sequencing. Nine probands (69.2%) were assigned a genetic diagnosis, with variants in MYRF, TMEM98, MFRP, and PRSS56. Two of four PRSS56 families harbored the previously described c.1066dupC variant implicated in over half of all reported PRSS56 kindreds, with different surrounding haplotypes in each family suggesting a mutational hotspot. Individuals with a genetic diagnosis had shorter mean axial lengths and higher hyperopia than those without, with recessive forms associated with the most extreme phenotypes. These findings detail the genetic architecture of nanophthalmos and posterior microphthalmos in a cohort of predominantly European ancestry, their relative clinical phenotypes, and highlight the shared genetic architecture of rare and common disorders of refractive error.


Subject(s)
Glaucoma, Angle-Closure/genetics , Hyperopia/genetics , Membrane Proteins/genetics , Microphthalmos/genetics , Serine Proteases/genetics , Transcription Factors/genetics , Australia/epidemiology , Cohort Studies , Eye/pathology , Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/pathology , Female , Frameshift Mutation/genetics , Glaucoma, Angle-Closure/pathology , Humans , Hyperopia/pathology , Male , Microphthalmos/pathology , Pedigree
12.
Hum Mol Genet ; 26(2): 438-453, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28073927

ABSTRACT

Primary open-angle glaucoma (POAG), the most common optic neuropathy, is a heritable disease. Siblings of POAG cases have a ten-fold increased risk of developing the disease. Intraocular pressure (IOP) and optic nerve head characteristics are used clinically to predict POAG risk. We conducted a genome-wide association meta-analysis of IOP and optic disc parameters and validated our findings in multiple sets of POAG cases and controls. Using imputation to the 1000 genomes (1000G) reference set, we identified 9 new genomic regions associated with vertical cup-disc ratio (VCDR) and 1 new region associated with IOP. Additionally, we found 5 novel loci for optic nerve cup area and 6 for disc area. Previously it was assumed that genetic variation influenced POAG either through IOP or via changes to the optic nerve head; here we present evidence that some genomic regions affect both IOP and the disc parameters. We characterized the effect of the novel loci through pathway analysis and found that pathways involved are not entirely distinct as assumed so far. Further, we identified a novel association between CDKN1A and POAG. Using a zebrafish model we show that six6b (associated with POAG and optic nerve head variation) alters the expression of cdkn1a. In summary, we have identified several novel genes influencing the major clinical risk predictors of POAG and showed that genetic variation in CDKN1A is important in POAG risk.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Glaucoma, Open-Angle/genetics , Homeodomain Proteins/genetics , Optic Nerve Diseases/genetics , Zebrafish Proteins/genetics , Female , Genome, Human , Genome-Wide Association Study , Glaucoma, Open-Angle/pathology , Humans , Intraocular Pressure/genetics , Male , Middle Aged , Optic Disk/pathology , Optic Nerve Diseases/pathology , Tonometry, Ocular
13.
Mol Vis ; 25: 527-534, 2019.
Article in English | MEDLINE | ID: mdl-31700225

ABSTRACT

Purpose: Nanophthalmos is a rare subtype of microphthalmia associated with high hyperopia and an increased risk of angle-closure glaucoma. We investigated the genetic cause of nanophthalmos and high hyperopia in an autosomal dominant kindred. Methods: A proband with short axial length, high hyperopia, and dextrocardia was subjected to exome sequencing. Human and rodent gene expression data sets were used to investigate the expression of relevant genes. Results: We identified a segregating heterozygous frameshift variant at the 3' end of the penultimate exon of MYRF. Using Myc-MYRF chromatin immunoprecipitation data from rat oligodendrocytes, MYRF was found to bind immediately upstream of the transcriptional start site of Tmem98, a gene that itself has been implicated in autosomal dominant nanophthalmos. MYRF and TMEM98 were found to be expressed in the human retina, with a similar pattern of expression across several dissected human eye tissues. Conclusions: C-terminal variants in MYRF, which are expected to escape nonsense-mediated decay, represent a rare cause of autosomal dominant nanophthalmos with or without dextrocardia or congenital diaphragmatic hernia.


Subject(s)
Eye Diseases, Hereditary/complications , Eye Diseases, Hereditary/genetics , Frameshift Mutation/genetics , Glaucoma, Angle-Closure/complications , Glaucoma, Angle-Closure/genetics , Hyperopia/complications , Hyperopia/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microphthalmos/complications , Microphthalmos/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Adult , Amino Acid Sequence , Animals , Base Sequence , Female , Genes, Dominant , Humans , Male , Membrane Proteins/metabolism , Pedigree , Rats , Transcription Factors/metabolism
14.
Ophthalmology ; 126(8): 1119-1130, 2019 08.
Article in English | MEDLINE | ID: mdl-30910584

ABSTRACT

PURPOSE: To investigate which clinical measures influence whether an individual demonstrates earliest glaucomatous structural progression on peripapillary retinal nerve fiber layer (pRNFL) or macular ganglion cell-inner plexiform layer (mGCIPL). DESIGN: Prospective, longitudinal cohort study. PARTICIPANTS: Two hundred seventy-one eyes from 207 individuals with statistically significant evidence of glaucomatous progression on OCT Guided Progression Analysis (GPA) software were drawn from a total of 1271 eyes from 686 individuals categorized as glaucoma suspect or having early manifest glaucoma undergoing glaucoma surveillance. METHODS: Individuals demonstrating earliest evidence of longitudinal progression on mGCIPL GPA event analysis were compared with individuals demonstrating evidence of earliest longitudinal progression on pRNFL GPA event analysis. MAIN OUTCOME MEASURES: Correlation of OCT event change analysis with intraocular pressure (IOP), clinical variables, and baseline thickness of the pRNFL and mGCIPL. RESULTS: Intraocular pressure, baseline pRNFL thickness, baseline mGCIPL thickness, and systemic hypertension were associated with location of first progression. Eyes demonstrating earliest longitudinal progression on mGCIPL had significantly lower maximum-recorded pretreatment IOP (mean difference, 3.90 mmHg; 95% confidence interval [CI], 2.37-5.43 mmHg; P < 0.001). The interval between progression on pRNFL and progression on mGCIPL increased by 12.4 months for every 5-mmHg increase in IOP (95% CI, 10.32-15.72 months). Eyes demonstrating earliest longitudinal progression on mGCIPL showed significantly lower baseline average pRNFL thickness than eyes progressing on pRNFL first (mean difference, 7.07 µm; 95% CI, 4.38-9.77 µm; P < 0.001). Eyes progressing first on mGCIPL parameters were 3.03 times more likely to demonstrate a new paracentral field defect than eyes progressing first on pRNFL parameters (odds ratio, 3.03; 95% CI, 1.26-7.28; P = 0.01). CONCLUSIONS: Clinical features, particularly pretreatment IOP, influence whether structural glaucoma progression is detected earlier with mGCIPL or pRNFL imaging. These data support the usefulness of mGCIPL imaging in addition to pRNFL analysis for detection of glaucoma progression, particularly in patients with normal IOP.


Subject(s)
Glaucoma/physiopathology , Intraocular Pressure/physiology , Macula Lutea/pathology , Nerve Fibers/pathology , Retinal Ganglion Cells/pathology , Aged , Disease Progression , Female , Glaucoma/diagnosis , Humans , Longitudinal Studies , Male , Middle Aged , Prospective Studies
15.
Mol Vis ; 24: 261-273, 2018.
Article in English | MEDLINE | ID: mdl-29618921

ABSTRACT

Purpose: Aniridia is a congenital disorder caused by variants in the PAX6 gene. In this study, we assessed the involvement of PAX6 in patients with aniridia from Australasia and Southeast Asia. Methods: Twenty-nine individuals with aniridia from 18 families originating from Australia, New Caledonia, Cambodia, Sri Lanka, and Bhutan were included. The PAX6 gene was investigated for sequence variants and analyzed for deletions with multiplex ligation-dependent probe amplification. Results: We identified 11 sequence variants and six chromosomal deletions, including one in mosaic. Four deleterious sequence variants were novel: p.(Pro81HisfsTer12), p.(Gln274Ter), p.(Ile29Thr), and p.(Met1?). Ocular complications were associated with a progressive loss of visual function as shown by a visual acuity ≤ 1.00 logMAR reported in 65% of eyes. The prevalence of keratopathy was statistically significantly higher in the Australasian cohort (78.6%) compared with the Southeast Asian cohort (9.1%, p=0.002). Variants resulting in protein truncating codons displayed limited genotype-phenotype correlations compared with other variants. Conclusions: PAX6 variants and deletions were identified in 94% of patients with aniridia from Australasia and Southeast Asia. This study is the first report of aniridia and variations in PAX6 in individuals from Cambodia, Sri Lanka, Bhutan, and New Caledonia, and the largest cohort from Australia.


Subject(s)
Aniridia/genetics , Chromosome Deletion , Genetic Association Studies , Mosaicism , PAX6 Transcription Factor/genetics , Adolescent , Adult , Aniridia/diagnosis , Aniridia/pathology , Asia, Southeastern , Australasia , Base Sequence , Child , Cohort Studies , Exons , Female , Gene Expression , Genotype , Humans , Inheritance Patterns , Introns , Male , Middle Aged , PAX6 Transcription Factor/deficiency , Pedigree , Phenotype
16.
Mol Vis ; 24: 478-484, 2018.
Article in English | MEDLINE | ID: mdl-30090012

ABSTRACT

Purpose: Inherited retinal dystrophies are a clinically and genetically heterogeneous group of disorders. Molecular diagnosis has proven utility for affected individuals. In this study, we report an individual enrolled in the Australian Inherited Retinal Disease Registry and DNA Bank diagnosed with clinical features overlapping between Leber congenital amaurosis and retinitis pigmentosa. Methods: DNA from the proband was sequenced using a gene panel for inherited retinal disorders, and a single nucleotide polymorphism (SNP) array was conducted to detect the presence of deletions and uniparental disomy. Results: We identified a novel homozygous variant (c.524dupC, p.(Pro176ThrfsTer7)) in TULP1 resulting from maternal uniparental isodisomy of chromosome 6. The patient had clinical features consistent with biallelic pathogenic variants in TULP1, including congenital nystagmus, night blindness, non-recordable electroretinogram, mild myopia, and mild peripheral pigmentary changes in the fundus. Conclusions: This is the first report of uniparental disomy 6 and a homozygous variant in TULP1 associated with a rod-cone dystrophy. Molecular diagnosis of inherited retinal dystrophies is essential to inform the mode of transmission and clinical management, and to identify potential candidates for future gene-specific therapies.


Subject(s)
Eye Proteins/genetics , Leber Congenital Amaurosis/genetics , Myopia/genetics , Night Blindness/genetics , Nystagmus, Congenital/genetics , Retinitis Pigmentosa/genetics , Uniparental Disomy , Chromosomes, Human, Pair 6/chemistry , Electroretinography , Female , Gene Expression , Homozygote , Humans , Leber Congenital Amaurosis/diagnosis , Leber Congenital Amaurosis/pathology , Maternal Inheritance , Mutation , Myopia/diagnosis , Myopia/pathology , Night Blindness/diagnosis , Night Blindness/pathology , Nystagmus, Congenital/diagnosis , Nystagmus, Congenital/pathology , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/pathology , Young Adult
17.
BMC Med Genet ; 18(1): 52, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28482824

ABSTRACT

BACKGROUND: Cataract is a major cause of severe visual impairment in childhood. The purpose of this study was to determine the genetic cause of syndromic congenital cataract in an Australian mother and son. METHOD: Fifty-one genes associated with congenital cataract were sequenced in the proband using a custom Ampliseq library on the Ion Torrent Personal Genome Machine (PGM). Reads were aligned against the human genome (hg19) and variants were annotated. Variants were prioritised for validation by Sanger sequencing if they were novel, rare or previously reported to be associated with paediatric cataract and were predicted to be protein changing. Variants were assessed for segregation with the phenotype in the affected mother. RESULT: A novel likely pathogenic variant was identified in the transactivation domain of the MAF gene (c.176C > G, p.(Pro59Arg)) in the proband and his affected mother., but was absent in 326 unrelated controls and absent from public variant databases. CONCLUSION: The MAF variant is the likely cause of the congenital cataract, Asperger syndrome, seizures, hearing loss and facial characteristics in the proband, providinga diagnosis of Aymé-Gripp syndrome for the family.


Subject(s)
Cataract/congenital , Developmental Disabilities/genetics , Hearing Loss/genetics , Maf Transcription Factors/genetics , Mutation, Missense , Seizures/genetics , Adult , Amino Acid Sequence , Animals , Cataract/genetics , Female , Humans , Maf Transcription Factors/chemistry , Male , Pedigree , Sequence Homology, Amino Acid , Young Adult
18.
Ophthalmology ; 124(3): 303-309, 2017 03.
Article in English | MEDLINE | ID: mdl-27993484

ABSTRACT

PURPOSE: To assess the difference in severity of disease in primary open-angle glaucoma (POAG) patients with a Myocilin (MYOC) disease-causing variant who presented through normal clinical pathways (Clinical cases) versus those who were examined following genetic testing (Genetic cases). DESIGN: Retrospective clinical and molecular study. PARTICIPANTS: Seventy-three MYOC mutation carriers identified through the Australian and New Zealand Registry of Advanced Glaucoma. METHODS: Individuals were classified based on how they first presented to an ophthalmologist: Clinical cases were referred by their general practitioner or optometrist, and Genetic cases were referred following positive results from genetic testing for the previously identified familial MYOC variant (cascade genetic testing). All cases were then sub-classified into 4 groups (unaffected, glaucoma suspect, glaucoma, advanced glaucoma) according to the severity of disease at the time of their first examination by an ophthalmologist. MAIN OUTCOME MEASURES: Glaucoma clinical parameters and age at presentation. RESULTS: At their first examination, 83% of Genetic cases were unaffected and 17% were glaucoma suspect, whereas among Clinical cases 44% were glaucoma suspect, 28% had glaucoma, and 28% had advanced glaucoma. Genetic cases were significantly younger at presentation than Clinical cases (40.6±12.5 vs. 47.5±16.7 years; P = 0.018). The mean highest intraocular pressure (32.2±9.7 vs. 17.6±3.6 mmHg; P < 0.001), cup-to-disc ratio (0.65±0.27 vs. 0.48±0.13; P = 0.006), and mean deviation on visual field testing (-10.0±10.3 vs. -1.2±1.2; P < 0.001) were all significantly worse in Clinical cases compared with Genetic cases. Individuals with common MYOC p.Gln368Ter variant were further analyzed separately to account for the phenotypic variability of different disease-causing variants. All findings remained significant after adjusting for the common MYOC p.Gln368Ter variant. CONCLUSIONS: Our findings demonstrated that MYOC cascade genetic testing for POAG allows identification of at-risk individuals at an early stage or even before signs of glaucoma are present. To our knowledge, this is the first study to demonstrate the clinical utility of predictive genetic testing for MYOC glaucoma.


Subject(s)
Cytoskeletal Proteins/genetics , DNA Mutational Analysis , Eye Proteins/genetics , Glaucoma, Open-Angle/diagnosis , Glaucoma, Open-Angle/genetics , Glycoproteins/genetics , Mutation , Adolescent , Adult , Aged , Aged, 80 and over , Female , Genetic Testing , Humans , Intraocular Pressure , Male , Middle Aged , Ocular Hypertension/diagnosis , Ocular Hypertension/genetics , Physical Examination , Polymerase Chain Reaction , Retrospective Studies , Risk Factors
19.
BMC Med Genet ; 17: 30, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27080696

ABSTRACT

BACKGROUND: Glaucoma is a leading cause of irreversible blindness. Pathogenic variants in the Myocilin gene (MYOC) cause juvenile open angle glaucoma (JOAG) in 8-36% of cases, and display an autosomal dominant inheritance with high penetrance. Molecular diagnosis is important for early identification as therapies are effective in minimizing vision loss and MYOC variants can be associated to severe glaucoma. MYOC variants are usually inherited, however a fifth of carriers do not report a family history. The occurrence of de novo MYOC variants is currently unknown. CASE PRESENTATION: In this study we investigated a 14 year old male Caucasian patient diagnosed with JOAG, and no family history of glaucoma. A novel probably deleterious MYOC:p.(Pro254Leu) variant was identified in the index case. This variant was not present in the parents or the siblings. CONCLUSION: This is the second report of a de novo MYOC variant in a sporadic case of JOAG and it is currently unknown if this mechanism occurs more frequently. This finding emphasizes the importance of screening individuals with JOAG for MYOC mutations irrespective of a negative family history.


Subject(s)
Cytoskeletal Proteins/genetics , Eye Proteins/genetics , Genetic Variation , Glaucoma, Open-Angle/genetics , Glycoproteins/genetics , Adolescent , Amino Acid Sequence , Cytoskeletal Proteins/metabolism , Eye Proteins/metabolism , Genetic Testing , Glaucoma, Open-Angle/diagnosis , Glycoproteins/metabolism , Humans , Male , Molecular Sequence Data , Pedigree , White People/genetics
20.
Mol Vis ; 21: 160-4, 2015.
Article in English | MEDLINE | ID: mdl-25750510

ABSTRACT

PURPOSE: To evaluate the prevalence and the diagnostic utility of testing for CYP1B1 copy number variation (CNV) in primary congenital glaucoma (PCG) cases unexplained by CYP1B1 point mutations in The Australian and New Zealand Registry of Advanced Glaucoma. METHODS: In total, 50 PCG cases either heterozygous for disease-causing variants or with no CYP1B1 sequence variants were included in the study. CYP1B1 CNV was analyzed by Multiplex Ligation-dependent Probe Amplification (MLPA). RESULTS: No deletions or duplications were found in any of the cases. CONCLUSION: This is the first study to report on CYP1B1 CNV in PCG cases. Our findings show that this mechanism is not a major contributor to the phenotype and is of limited diagnostic utility.


Subject(s)
Cytochrome P-450 CYP1B1/genetics , Gene Dosage , Glaucoma/genetics , Child, Preschool , Female , Gene Expression , Genetic Variation , Genotype , Glaucoma/congenital , Glaucoma/pathology , Heterozygote , Humans , Male , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL