Ophthalmology (Eye & Ear Hospital) - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 76
  • Item
    Thumbnail Image
    AAV-Mediated CRISPR/Cas Gene Editing of Retinal Cells In Vivo
    Hung, SSC ; Chrysostomou, V ; Li, F ; Lim, JKH ; Wang, J-H ; Powell, JE ; Tu, L ; Daniszewski, M ; Lo, C ; Wong, RC ; Crowston, JG ; Pebay, A ; King, AE ; Bui, BV ; Liu, G-S ; Hewitt, AW (ASSOC RESEARCH VISION OPHTHALMOLOGY INC, 2016-06)
    PURPOSE: Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein (Cas) has recently been adapted to enable efficient editing of the mammalian genome, opening novel avenues for therapeutic intervention of inherited diseases. In seeking to disrupt yellow fluorescent protein (YFP) in a Thy1-YFP transgenic mouse, we assessed the feasibility of utilizing the adeno-associated virus 2 (AAV2) to deliver CRISPR/Cas for gene modification of retinal cells in vivo. METHODS: Single guide RNA (sgRNA) plasmids were designed to target YFP, and after in vitro validation, selected guides were cloned into a dual AAV system. One AAV2 construct was used to deliver Streptococcus pyogenes Cas9 (SpCas9), and the other delivered sgRNA against YFP or LacZ (control) in the presence of mCherry. Five weeks after intravitreal injection, retinal function was determined using electroretinography, and CRISPR/Cas-mediated gene modifications were quantified in retinal flat mounts. RESULTS: Adeno-associated virus 2-mediated in vivo delivery of SpCas9 with sgRNA targeting YFP significantly reduced the number of YFP fluorescent cells of the inner retina of our transgenic mouse model. Overall, we found an 84.0% (95% confidence interval [CI]: 81.8-86.9) reduction of YFP-positive cells in YFP-sgRNA-infected retinal cells compared to eyes treated with LacZ-sgRNA. Electroretinography profiling found no significant alteration in retinal function following AAV2-mediated delivery of CRISPR/Cas components compared to contralateral untreated eyes. CONCLUSIONS: Thy1-YFP transgenic mice were used as a rapid quantifiable means to assess the efficacy of CRISPR/Cas-based retinal gene modification in vivo. We demonstrate that genomic modification of cells in the adult retina can be readily achieved by viral-mediated delivery of CRISPR/Cas.
  • Item
    Thumbnail Image
    Novel pleiotropic risk loci for melanoma and nevus density implicate multiple biological pathways
    Duffy, DL ; Zhu, G ; Li, X ; Sanna, M ; Iles, MM ; Jacobs, LC ; Evans, DM ; Yazar, S ; Beesley, J ; Law, MH ; Kraft, P ; Visconti, A ; Taylor, JC ; Lui, F ; Wright, MJ ; Henders, AK ; Bowdler, L ; Glass, D ; Ikram, AM ; Uitterlinden, AG ; Madden, PA ; Heath, AC ; Nelson, EC ; Green, AC ; Chanock, S ; Barrett, JH ; Brown, MA ; Hayward, NK ; MacGregor, S ; Sturm, RA ; Hewitt, AW ; Kayser, M ; Hunter, DJ ; Bishop, JAN ; Spector, TD ; Montgomery, GW ; Mackey, DA ; Smith, GD ; Nijsten, TE ; Bishop, DT ; Bataille, V ; Falchi, M ; Han, J ; Martins, NG (NATURE PUBLISHING GROUP, 2018-11-14)
    The total number of acquired melanocytic nevi on the skin is strongly correlated with melanoma risk. Here we report a meta-analysis of 11 nevus GWAS from Australia, Netherlands, UK, and USA comprising 52,506 individuals. We confirm known loci including MTAP, PLA2G6, and IRF4, and detect novel SNPs in KITLG and a region of 9q32. In a bivariate analysis combining the nevus results with a recent melanoma GWAS meta-analysis (12,874 cases, 23,203 controls), SNPs near GPRC5A, CYP1B1, PPARGC1B, HDAC4, FAM208B, DOCK8, and SYNE2 reached global significance, and other loci, including MIR146A and OBFC1, reached a suggestive level. Overall, we conclude that most nevus genes affect melanoma risk (KITLG an exception), while many melanoma risk loci do not alter nevus count. For example, variants in TERC and OBFC1 affect both traits, but other telomere length maintenance genes seem to affect melanoma risk only. Our findings implicate multiple pathways in nevogenesis.
  • Item
    Thumbnail Image
    Rare variants in optic disc area gene CARD10 enriched in primary open-angle glaucoma
    Zhou, T ; Souzeau, E ; Sharma, S ; Siggs, OM ; Goldberg, I ; Healey, PR ; Graham, S ; Hewitt, AW ; Mackey, DA ; Casson, RJ ; Landers, J ; Mills, R ; Ellis, J ; Leo, P ; Brown, MA ; MacGregor, S ; Burdon, KP ; Craig, JE (WILEY, 2016-11)
    BACKGROUND: Genome-wide association studies (GWAS) have identified association of common alleles with primary open-angle glaucoma (POAG) and its quantitative endophenotypes near numerous genes. This study aims to determine whether rare pathogenic variants in these disease-associated genes contribute to POAG. METHODS: Participants fulfilled strict inclusion criteria of advanced POAG at a young age of diagnosis. Myocilin mutation carriers were excluded using direct sequencing. Whole exome sequencing was performed on 187 glaucoma cases and 103 local screened nonglaucoma controls then joint-called with exomes of 993 previously sequenced Australian controls. GWAS-associated genes were assessed for enrichment of rare predicted pathogenic variants in POAG. Significantly enriched genes were compared against Exome Aggregation Consortium (ExAC) public control. RESULTS: Eighty-six GWAS disease or trait-associated glaucoma genes were captured and sequenced. CARD10 showed enrichment after Bonferroni correction for rare variants in glaucoma cases (OR = 13.2, P = 6.94 × 10-5) with mutations identified in 4.28% of our POAG cohort compared to 0.27% in controls. CARD10 was significantly associated with optic disc parameters in previous GWAS. The whole GWAS gene set showed no enrichment in POAG overall (OR = 1.12, P = 0.51). CONCLUSION: We report here an enrichment of rare predicted pathogenic coding variants within a GWAS-associated locus in POAG (CARD10). These findings indicate that both common and rare pathogenic coding variants in CARD10 may contribute to POAG pathogenesis.
  • Item
    Thumbnail Image
    Novel pleiotropic risk loci for melanoma and nevus density implicate multiple biological pathways (vol 9, 4774, 2018)
    Duffy, DL ; Zhu, G ; Li, X ; Sanna, M ; Iles, MM ; Jacobs, LC ; Evans, DM ; Yazar, S ; Beesley, J ; Law, MH ; Kraft, P ; Visconti, A ; Taylor, JC ; Liu, F ; Wright, MJ ; Henders, AK ; Bowdler, L ; Glass, D ; Ikram, MA ; Uitterlinden, AG ; Madden, PA ; Heath, AC ; Nelson, EC ; Green, AC ; Chanock, S ; Barrett, JH ; Brown, MA ; Hayward, NK ; MacGregor, S ; Sturm, RA ; Hewitt, AW ; Kayser, M ; Hunter, DJ ; Bishop, JAN ; Spector, TD ; Montgomery, GW ; Mackey, DA ; Smith, GD ; Nijsten, TE ; Bishop, DT ; Bataille, V ; Falchi, M ; Han, J ; Martin, NG ; Lee, JE ; Brossard, M ; Moses, EK ; Song, F ; Kumar, R ; Easton, DF ; Pharoah, PDP ; Swerdlow, AJ ; Kypreou, KP ; Harland, M ; Randerson-Moor, J ; Akslen, LA ; Andresen, PA ; Avril, M-F ; Azizi, E ; Scarra, GB ; Brown, KM ; Debniak, T ; Elder, DE ; Fang, S ; Friedman, E ; Galan, P ; Ghiorzo, P ; Gillanders, EM ; Goldstein, AM ; Gruis, NA ; Hansson, J ; Helsing, P ; Hocevar, M ; Hoiom, V ; Ingvar, C ; Kanetsky, PA ; Chen, WV ; Landi, MT ; Lang, J ; Lathrop, GM ; Lubinski, J ; Mackie, RM ; Mann, GJ ; Molven, A ; Novakovic, S ; Olsson, H ; Puig, S ; Puig-Butille, JA ; Radford-Smith, GL ; van der Stoep, N ; van Doorn, R ; Whiteman, DC ; Craig, JE ; Schadendorf, D ; Simms, LA ; Burdon, KP ; Nyholt, DR ; Pooley, KA ; Orr, N ; Stratigos, AJ ; Cust, AE ; Ward, SV ; Schulze, H-J ; Dunning, AM ; Demenais, F ; Amos, CI (NATURE PUBLISHING GROUP, 2019-01-14)
    The original version of this Article contained errors in the spelling of the authors Fan Liu and M. Arfan Ikram, which were incorrectly given as Fan Lui and Arfan M. Ikram. In addition, the original version of this Article also contained errors in the author affiliations which are detailed in the associated Publisher Correction.
  • Item
    Thumbnail Image
    Testosterone Pathway Genetic Polymorphisms in Relation to Primary Open-Angle Glaucoma: analysis in Two Large Datasets
    Bailey, JNC ; Gharahkhani, P ; Kang, JH ; Butkiewicz, M ; Sullivan, DA ; Weinreb, RN ; Aschard, H ; Allingham, RR ; Ashley-Koch, A ; Lee, RK ; Moroi, SE ; Brilliant, MH ; Wollstein, G ; Schuman, JS ; Fingert, JH ; Budenz, DL ; Realini, T ; Gaasterland, T ; Scott, WK ; Singh, K ; Sit, AJ ; Igo, RP ; Song, YE ; Hark, L ; Ritch, R ; Rhee, DJ ; Vollrath, D ; Zack, DJ ; Medeiros, F ; Vajaranant, TS ; Chasman, DI ; Christen, WG ; Pericak-Vance, MA ; Liu, Y ; Kraft, P ; Richards, JE ; Rosner, BA ; Hauser, MA ; Craig, JE ; Burdon, KP ; Hewitt, AW ; Mackey, DA ; Haines, JL ; MacGregor, S ; Wiggs, JL ; Pasquale, LR (ASSOC RESEARCH VISION OPHTHALMOLOGY INC, 2018-02)
    PURPOSE: Sex hormones may be associated with primary open-angle glaucoma (POAG), although the mechanisms are unclear. We previously observed that gene variants involved with estrogen metabolism were collectively associated with POAG in women but not men; here we assessed gene variants related to testosterone metabolism collectively and POAG risk. METHODS: We used two datasets: one from the United States (3853 cases and 33,480 controls) and another from Australia (1155 cases and 1992 controls). Both datasets contained densely called genotypes imputed to the 1000 Genomes reference panel. We used pathway- and gene-based approaches with Pathway Analysis by Randomization Incorporating Structure (PARIS) software to assess the overall association between a panel of single nucleotide polymorphisms (SNPs) in testosterone metabolism genes and POAG. In sex-stratified analyses, we evaluated POAG overall and POAG subtypes defined by maximum IOP (high-tension [HTG] or normal tension glaucoma [NTG]). RESULTS: In the US dataset, the SNP panel was not associated with POAG (permuted P = 0.77), although there was an association in the Australian sample (permuted P = 0.018). In both datasets, the SNP panel was associated with POAG in men (permuted P ≤ 0.033) and not women (permuted P ≥ 0.42), but in gene-based analyses, there was no consistency on the main genes responsible for these findings. In both datasets, the testosterone pathway association with HTG was significant (permuted P ≤ 0.011), but again, gene-based analyses showed no consistent driver gene associations. CONCLUSIONS: Collectively, testosterone metabolism pathway SNPs were consistently associated with the high-tension subtype of POAG in two datasets.
  • Item
    Thumbnail Image
    Utility of Self-Destructing CRISPR/Cas Constructs for Targeted Gene Editing in the Retina
    Li, F ; Hung, SSC ; Mohd Khalid, MKN ; Wang, J-H ; Chrysostomou, V ; Wong, VHY ; Singh, V ; Wing, K ; Tu, L ; Bender, JA ; Pebay, A ; King, AE ; Cook, AL ; Wong, RCB ; Bui, BV ; Hewitt, AW ; Liu, G-S (MARY ANN LIEBERT, INC, 2019-11-01)
    Safe delivery of CRISPR/Cas endonucleases remains one of the major barriers to the widespread application of in vivo genome editing. We previously reported the utility of adeno-associated virus (AAV)-mediated CRISPR/Cas genome editing in the retina; however, with this type of viral delivery system, active endonucleases will remain in the retina for an extended period, making genotoxicity a significant consideration in clinical applications. To address this issue, we have designed a self-destructing "kamikaze" CRISPR/Cas system that disrupts the Cas enzyme itself following expression. Four guide RNAs (sgRNAs) were initially designed to target Streptococcus pyogenes Cas9 (SpCas9) and after in situ validation, the selected sgRNAs were cloned into a dual AAV vector. One construct was used to deliver SpCas9 and the other delivered sgRNAs directed against SpCas9 and the target locus (yellow fluorescent protein [YFP]), in the presence of mCherry. Both constructs were packaged into AAV2 vectors and intravitreally administered in C57BL/6 and Thy1-YFP transgenic mice. After 8 weeks, the expression of SpCas9 and the efficacy of YFP gene disruption were quantified. A reduction of SpCas9 mRNA was found in retinas treated with AAV2-mediated YFP/SpCas9 targeting CRISPR/Cas compared with those treated with YFP targeting CRISPR/Cas alone. We also show that AAV2-mediated delivery of YFP/SpCas9 targeting CRISPR/Cas significantly reduced the number of YFP fluorescent cells among mCherry-expressing cells (∼85.5% reduction compared with LacZ/SpCas9 targeting CRISPR/Cas) in the transfected retina of Thy1-YFP transgenic mice. In conclusion, our data suggest that a self-destructive "kamikaze" CRISPR/Cas system can be used as a robust tool for genome editing in the retina, without compromising on-target efficiency.
  • Item
    Thumbnail Image
    Response: Cycloplegia in refraction: age and cycloplegics
    Sanfilippo, PG ; Chu, B-S ; Bigault, O ; Kearns, LS ; Boon, M-Y ; Young, TL ; Hammond, CJ ; Hewitt, AW ; Mackey, DA (WILEY-BLACKWELL, 2016-08)
  • Item
    Thumbnail Image
    A single-cell transcriptome atlas of the adult human retina
    Lukowski, SW ; Lo, CY ; Sharov, AA ; Nguyen, Q ; Fang, L ; Hung, SSC ; Zhu, L ; Zhang, T ; Grunert, U ; Nguyen, T ; Senabouth, A ; Jabbari, JS ; Welby, E ; Sowden, JC ; Waugh, HS ; Mackey, A ; Pollock, G ; Lamb, TD ; Wang, P-Y ; Hewitt, AW ; Gillies, MC ; Powell, JE ; Wong, RCB (WILEY, 2019-09-16)
    The retina is a specialized neural tissue that senses light and initiates image processing. Although the functional organization of specific retina cells has been well studied, the molecular profile of many cell types remains unclear in humans. To comprehensively profile the human retina, we performed single-cell RNA sequencing on 20,009 cells from three donors and compiled a reference transcriptome atlas. Using unsupervised clustering analysis, we identified 18 transcriptionally distinct cell populations representing all known neural retinal cells: rod photoreceptors, cone photoreceptors, Müller glia, bipolar cells, amacrine cells, retinal ganglion cells, horizontal cells, astrocytes, and microglia. Our data captured molecular profiles for healthy and putative early degenerating rod photoreceptors, and revealed the loss of MALAT1 expression with longer post-mortem time, which potentially suggested a novel role of MALAT1 in rod photoreceptor degeneration. We have demonstrated the use of this retina transcriptome atlas to benchmark pluripotent stem cell-derived cone photoreceptors and an adult Müller glia cell line. This work provides an important reference with unprecedented insights into the transcriptional landscape of human retinal cells, which is fundamental to understanding retinal biology and disease.
  • Item
    Thumbnail Image
    Seeing the impact of the Glaucoma Inheritance Study in Tasmania after 25 years
    Mackey, DA ; Craig, JE ; Hewitt, AW (WILEY, 2019-07)
  • Item
    Thumbnail Image
    Current state and future prospects of artificial intelligence in ophthalmology: a review
    Hogarty, DT ; Mackey, DA ; Hewitt, AW (WILEY, 2019-01)
    Artificial intelligence (AI) has emerged as a major frontier in computer science research. Although AI has broad application across many medical fields, it will have particular utility in ophthalmology and will dramatically change the diagnostic and treatment pathways for many eye conditions such as corneal ectasias, glaucoma, age-related macular degeneration and diabetic retinopathy. However, given that AI has primarily been driven as a computer science, its concepts and terminology are unfamiliar to many medical professionals. Important key terms such as machine learning and deep learning are often misunderstood and incorrectly used interchangeably. This article presents an overview of AI and new developments relevant to ophthalmology.