ARVO 2015 Annual Meeting Abstracts 372 Stem cell biology Tuesday, May 05, 2015 3:45 PM–5:30 PM Exhibit Hall Poster Session Program #/Board # Range: 3572–3588/A0001–A0017 Organizing Section: Retinal Cell Biology Program Number: 3572 Poster Board Number: A0001 Presentation Time: 3:45 PM–5:30 PM Generation of neuroretina from induced pluripotent stem cells and effects of Sonic Hedgehog Camille Yvon, Conor Ramsden, Amanda-Jayne F. Carr, Matthew Smart, Michael Powner, Amelia Lane, Britta Nommitse, Lyndon da Cruz, Pete Coffey. Institute of Ophthalmology, UCL, London, United Kingdom. Purpose: Induced pluripotent stem cells (iPSCs) have the potential to provide robust models for the study of human retinogenesis and treatment therapies for retinal diseases. iPSC differentiation into neural retina (NR) can be driven in vitro by lineage-determining transcription factors. However, there exists a great source of variability between published protocols. We sought to determine if iPSCs could generate optic vesicle like structures or NR, by varying culture conditions of Matrigel™ and Sonic Hedgehog (Shh). Methods: We compared culture conditions for NR differentiation using two batches of Matrigel™ (M1 and M2), and variable concentrations of Shh trapped in the Matrigel coating the plate. Early changes in retinal development were monitored by immunocytochemistry and quantitative polymerase chain reaction. Results: We report the generation of optic vesicle and cup structures from iPSCs, using our modified protocol. It was found that embryoid bodies cultured in M2 showed higher expression of retinal development association transcription factors such as Pax6, Rax, Otx2 and Chx10 compared to M1. Embryoid bodies cultured in elevated Shh concentrations induced a greater percentage of organized structures and increased expression of eye field transcription factors. Conclusions: Our modified protocol using Shh delivered via plate coating provides practical and efficient ways of generating NR from iPSCs, thus helping to optimise the differentiation protocol. Commercial Relationships: Camille Yvon, None; Conor Ramsden, None; Amanda-Jayne F. Carr, None; Matthew Smart, None; Michael Powner, None; Amelia Lane, None; Britta Nommitse, None; Lyndon da Cruz, None; Pete Coffey, None Program Number: 3573 Poster Board Number: A0002 Presentation Time: 3:45 PM–5:30 PM WNT signaling during patterning of human iPSC-derived optic vesicle-like structures Anna Petelinsek1, Lynda S. Wright1, Elizabeth E. Capowski1, Jessica Lien1, Isabel Pinilla Lozano2, 5, Jishnu Saha1, Joe Phillips1, David M. Gamm3, 4. 1University of Wisconsin-Madison, Madison, WI; 2 Ophthalmology, University Hospital Lozano Blesa, Zaragoza, Spain; 3 Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI; 4McPherson Eye Research Institute, Madison, WI; 5Aragon Health Sciences Institute, IIS Aragon, Zaragoza, Spain. Purpose: VSX2, a homeodomain transcription factor expressed in neural retina progenitor cells (NRPCs), has a prominent role in optic vesicle (OV) patterning. Recently, we used hiPSCs to demonstrate that an R200Q mutation in the DNA binding region of VSX2 caused a partial cell fate switch from NR to RPE. RNAseq data comparing NR from wild type (WT) and (R200Q)VSX2 hiPSCs suggested that the WNT pathway was altered in VSX2 mutant OVs. To further investigate this finding, we examined the spatiotemporal expression of MITF, VSX2, and canonical WNT signaling proteins during hiPSC-OV patterning. Methods: hiPSC-OVs were generated from an individual bearing homozygous R200Q mutations in VSX2 and an unaffected sibling using our established methods. We performed ICC to localize expression of VSX2, MITF, and the WNT markers b-catenin, WLS, and LEF1 at days 14, 18, 35 and 50 of differentiation. Results: Retinal differentiation of WT hiPSCs resulted in induction of a MITF+ population of OV cells at d14, a subset of which initially coexpressed VSX2. Also at d14, expression of the WNT markers WLS and LEF1, along with nuclear localization of the canonical WNT pathway effector b-catenin, were detected in MITF+ cells but seldom in VSX2+ cells. By d18 of differentiation, WT OV cells exclusively expressed either MITF or VSX2, with the latter developing as NR and the former adopting an RPE fate. However, when (R200Q)VSX2 hiPSCs were differentiated toward a retinal fate, VSX2 and MITF remained coexpressed as late as d18. Nuclear localization of b-catenin was robustly observed in VSX2+/MITF+ cells at d14 in (R200Q)VSX2 OV cells, and WLS and LEF1 displayed aberrant coexpression with VSX2 as well. However, once NR and RPE were established, differences in canonical WNT signaling were not detected between WT and mutant. WLS and LEF1 were observed in maturing RPE but not NR in both WT and mutant OVs, and nuclear localization of b-catenin was absent in both by d35. Conclusions: We demonstrated that canonical WNT signaling proteins colocalized with MITF+ OV and RPE cells during early hiPSC-OV patterning, but were downregulated in NRPCs following expression of WT VSX2. Early NRPCs expressing mutant (R200Q) VSX2 displayed prolonged expression of MITF and WNT markers, along with nuclear localization of b-catenin. This data suggests that VSX2 DNA binding activity is associated with downregulation of canonical WNT signaling during NRPC specification. Commercial Relationships: Anna Petelinsek, None; Lynda S. Wright, None; Elizabeth E. Capowski, None; Jessica Lien, None; Isabel Pinilla Lozano, None; Jishnu Saha, None; Joe Phillips, None; David M. Gamm, Cellular Dynamics International (S) Support: This work was supported by National Institutes of Health grant numbers R01 EY21218, P30 HD03352; the Foundation Fighting Blindness Wynn-Gund Translation Research Award; the Retina Research Foundation (Kathryn and Latimer Murfee and Emmett A Humble Chairs); the Choroideremia Research Foundation; and the McPherson Eye Research Institute (Sandra Lemke Trout Chair in Eye Research). Program Number: 3574 Poster Board Number: A0003 Presentation Time: 3:45 PM–5:30 PM Neural Potential of Human Limbal Cells Xiaoli Chen1, Pawez Hossain1, 2, Helen Griffiths1, Jennifer Scott1, Andrew J. Lotery1, 2. 1Faculty of Medicine, University of Southampton, Southampton, United Kingdom; 2Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom. Purpose: Previous studies have shown that neural colonies (neurospheres) derived from adult mouse limbus are neural crest originated stem/progenitor cells, and can differentiate into functional neurons and/or cells expressing retinal specific markers in vitro. The aim of this study is to investigate whether limbal neurosphere cells (LNS) can be derived from aged human limbus, and differentiate into retinal like cells in vitro and in vivo. Methods: Human limbal tissues used in this study were from two resources: (1) aged donor eyes from Bristol Eye Bank (age ranged from 72-97), and (2) healthy limbal tissue from live patients who underwent conjunctival surgery (size 1 mm2, age ranged from 34- ©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission to reproduce any abstract, contact the ARVO Office at pubs@arvo.org. ARVO 2015 Annual Meeting Abstracts 85). Human limbus cells were isolated and cultured in the presence of mitogens. Following co-culture with developing retinal cells or in the presence of extrinsic factors, LNS and their progeny were characterized using immunocytochemistry and/or RT-PCR. Enhanced green fluorescent protein-tagged LNS were transplanted into the subretinal space of neonatal mice. The potential for limbal cells to differentiate into retinal like cells and integrate into the host retina was assessed by immunohistochemistry after 2-5 weeks. Results: Human LNS were successfully generated from aged donor limbal tissues through a serum free sphere forming assay. Human LNS expressed neural stem cell markers, including Sox2 (31.2 ± 10.2%) and Nestin (34.8 ± 2.2%). For the superficial limbal tissues (1 mm2) obtained from live patients, no apparent LNS were generated, but cells expressing Nestin (3-5%) and early differentiated neuronal marker beta-III tubulin (15-20%) can be grown through explant culture in the presence of serum and mitogens. Following co-culture with developing retinal cells or in presence of extrinsic factors, low levels of retinal progenitor markers, such as Lhx2, Pax6 and Rx were detected in human LNS at the transcription level. Mature photoreceptor specific markers were not observed in human LNS either in vitro or in vivo. Conclusions: Here we demonstrate that cells with neural potential can be derived from aged human limbal tissue or 1 mm2 of superficial limbal tissues from adult patients. Other approaches are needed to promote human limbal cells transdifferentiation into retinal lineage. However, their surgical accessibility and presence in aged individuals make them an attractive cell resource for autologous cell rescue of degenerative retinal diseases. Commercial Relationships: Xiaoli Chen, None; Pawez Hossain, None; Helen Griffiths, None; Jennifer Scott, None; Andrew J. Lotery, None Support: National Eye Research Centre (NERC), Rosetrees Trust, T.F.C. Frost Charity and the Gift of Sight Appeal. Program Number: 3575 Poster Board Number: A0004 Presentation Time: 3:45 PM–5:30 PM In vitro modeling of human retinogenesis with pluripotent stem cells Akshayalakshmi Sridhar1, Sarah Ohlemacher1, Jason S. Meyer1, 2 1 . Biology, Indiana Univ Purdue Univ Indianapolis, Indianapolis, IN; 2Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN. Purpose: Human pluripotent stem cells (hPSCs) provide a unique ability to study some of the earliest events of human development, particularly some of the earliest events in human retinogenesis such as the establishment of a definitive retinal fate from a more primitive neural progenitor source. In this role, hPSCs may provide an in vitro model for understanding the complex interplay of transcription factors involved in the acquisition of a retinal fate from an unspecified pluripotent cell population. Methods: hPSCs were differentiated as previously described and samples were collected every two days, starting from the undifferentiated state through when cells acquired either retinal or non-retinal forebrain identities. Immunocytochemistry and qRTPCR approaches were undertaken to identify candidate transcription involved in retinal fate establishment. Lentiviral-mediated overexpression and shRNA knockdown approaches assessed the role of candidate transcription factors in the specification of a retinal fate apart from other non-retinal neural fates. Furthermore, epigenetic approaches assessed the role of DNA methylation in retinal and forebrain fate determination. Results: Candidate transcription factors were identified underlying the establishment of a retinal fate apart from other neural lineages. Neural transcription factors including PAX6 and OTX2 were expressed early while retinal-associated transcription factors such as SIX6 were expressed at slightly later timepoints. Upon establishment of an anterior neural identity, expression patterns of certain transcription factors such as RAX became more restricted to subpopulations of cells, indicating the emergence of retinal and forebrain populations from the same primitive anterior neural population. Gene overexpression and knockdown experiments investigated the mechanism of action of these candidate transcription factors. Furthermore, epigenetic analysis demonstrated that DNA methylation could potentially account for differential gene expression in the establishment of retinal phenotypes apart from alternate neural lineages. Conclusions: Preliminary results begin to elucidate the complex interplay of transcription factors involved in the specification of a retinal fate from differentiating hPSCs. Overall, these results will help to better establish hPSCs as a valuable in vitro system with which to study some of the earliest events of human retinogenesis. Commercial Relationships: Akshayalakshmi Sridhar, None; Sarah Ohlemacher, None; Jason S. Meyer, None Support: NH Grant R01 EY024984-01, BrightFocus G2012027 Program Number: 3576 Poster Board Number: A0005 Presentation Time: 3:45 PM–5:30 PM Characterizing Sox2+cell in the adult mouse optic nerve lamina Yan Guo, Zara Mehrabyan, Steven L. Bernstein. Ophthalmology, Univ of Maryland Sch of Medicine, Baltimore, MD. Purpose: The optic nerve lamina (ONL) is a unique optic nerve structure bordering the retina and optic nerve. The ONL has a rich vasculature supply compared with rest of the optic nerve, and plays an important role in many optic nerve diseases. We recently found that there are abundant Sox2+ cells in the adult lamina. Sox2 is a nuclear transcription factor, essential for the pluripotency of adult neural stem cells (NSC) in central nervous system (CNS). We wanted to characterize these Sox2+ cells in order to have a better understanding of this unique region. Methods: 6 wild type mice (C57BL/6J) at age postnatal day 30-36 were utilized in the study. Mice were perfused with 4% paraformaldehyde. The optic laminae with approximately 0.5mm optic nerve were dissected and post fixed in PFA over night, and transferred to PBS, then 30% sucrose, and embedded in OCT and quickly frozen in dry ice. Ten micron thick frozen sections were prepared for immunohistochemical analysis. We performed immunohistochemistry using antibodies to Sox2, Nestin, GFAP, Ki67, Ng2, NeuN, Laminin. Slides were examined on an Olympus 900 laser confocal microscope. Results: Sox2+ cells are abundant in the ONL and fewer in the distal ON. In the retina, Sox2+ cells are located in the inner nuclear and ganglion cell layers. Sox2+ cells in ONL and ON are strongly associated with GFAP (glial astrocyte and neural stem cell marker), as well as nestin (neural stem cell marker). Some Sox2+ cells in the ONL are Ki67 positive. The Sox2 expressing cells in the ONL and their association with the other cell markers are very similar to those seen in the CNS subgranular zone (SGZ) in mice of same age. No association of Sox2+ and iba1 (macrophage marker) was seen, nor were Ng2 (oligodendrocyte precursor) or laminin (blood vessel). In the retina, Sox2+ cells are not clearly associated with GFAP, or nestin. They do not co-localize with NeuN (neuron marker) or Ki67. Conclusions: Sox2+ cells are abundantly expressed in the adult mouse ONL. Their expression pattern and their associations with other stem cell markers such as nestin, and mitotic markers such as KI67, are similar to what have seen in SGZ. Sox2+ cells in the retina do not associate with GFAP, implying that these cells may be of a ©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission to reproduce any abstract, contact the ARVO Office at pubs@arvo.org. ARVO 2015 Annual Meeting Abstracts different type. Collectively, these findings suggest that similar to the pluripotent neural stem cells seen in the SGZ, ONL Sox2+ cells may possess the ability to develop into other cell types. Commercial Relationships: Yan Guo, None; Zara Mehrabyan, None; Steven L. Bernstein, None Support: EY-015304 Program Number: 3577 Poster Board Number: A0006 Presentation Time: 3:45 PM–5:30 PM Scalable and reliable generation of retinal cells from human transgene-free induced pluripotent stem cells under defined xenofree and feeder-free conditions Olivier Goureau1, Amelie Slembrouck1, Angelique Terray1, Giuliana Gagliardi1, Celine Nanteau1, Jose A. Sahel1, 2, Sacha Reichman1. 1 Institut de la Vision, INSERM U968; Sorbonne Universités UPMCParis 06; CNRS UMR7210, Paris, France; 2Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, Paris, France. Purpose: For retinal cell therapy based on human induced pluripotent stem (iPS) cells, one of the major challenges is to develop essential culture conditions for the use of these cells for future clinical purposes. Until recently, iPS cell culture (maintenance and/or differentiation) has been carried out using feeder cells and/or culture media that contain animal products. Here, we adapted our new retinal differentiation method using confluent human iPS cells, bypassing cell clumps or embryoid body formation and in absence of Matrigel or serum (Reichman et al. PNAS 2014; 111:8518), in a well-defined xeno-free / feeder-free (XF/FF) system Methods: Integration-free iPS cells cultured on mouse embryonic fibroblasts were transferred onto vitronectin-coating plates and cultured with xeno-free medium. Confluent iPS cells obtained in these XF/FF conditions were directed toward a retinal lineage in a serum free proneural medium containing N2 supplement. Emergent neural retina (NR)-like structures were isolated and cultured in floating conditions for their maturation with a serum free proneural medium. Capacity for retinal differentiation was determined by immunohistochemistry and qRT-PCR analysis triggering specific developmental and mature retinal markers Results: In less than one month, confluent iPS cells are able to generate self-forming NR-like structures containing multipotent retinal progenitor cells (RPCs). Floating cultures of isolated neuroretinal tissue enabled the differentiation of RPCs into all types of retinal cells. Early-born retinal cells (i.e. ganglion, amacrine and horizontal cells) were identified after one month in culture, and lateborn retinal cells (i.e. photoreceptors, Muller glial and bipolar cells) started to appear after two months Conclusions: These data demonstrate that human iPS cell lines can be maintained and directed to differentiate into retinal cell types under XF/FF conditions that are required for translation to clinical applications. In this context the reliable generation of retinal ganglion cells and photoreceptor precursors could find important applications in regenerative medicine Commercial Relationships: Olivier Goureau, None; Amelie Slembrouck, None; Angelique Terray, None; Giuliana Gagliardi, None; Celine Nanteau, None; Jose A. Sahel, None; Sacha Reichman, None Support: ANR [GPiPS: ANR-2010-RFCS005]; [ANR-11IDEX-0004-02] in the frame of the LABEX LIFESENSES [ANR-10LABX-65]; Regional Council of Ile-de-France [DIM-Biothérapies]; SATT LUTECH Program Number: 3578 Poster Board Number: A0007 Presentation Time: 3:45 PM–5:30 PM Biasing early primitive ectoderm-like cells toward retinal cone photoreceptors Andrea S. Viczian1, 2, Kimberly A. Wong1, 2, Michael Trembley3. 1 Ophthalmology, Center for Vision Res, SUNY Upstate Medical Univ, Syracuse, NY; 2SUNY Eye Institute, Syracuse, NY; 3 Department of Pharmacology & Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY. Purpose: In the developing embryo, primitive ectoderm formation is lineage-restricted in response to extrinsic factors. The extrinsic factor, Noggin, plays a key role in inducing retinal cell markers in cultured human embryonic stem (ES) cells. In contrast, mouse ES cells do not express retinal markers when exposed to Noggin. Human ES cells have been shown to share more characteristics with mouse primitive ectoderm than with mouse ES cells. We conducted our study to determine if driving mouse ES cells to a primitive ectoderm lineage would allow them to respond to Noggin, and induce retinal cell markers. Methods: Mouse ES cells were treated with conditioned media and transformed into primitive ectoderm-like (EPL) cells. EPL-induced cells were then treated with Noggin and subsequently grown in differentiation media. Immunocytochemical and qPCR were used to characterize the cells. Results: If first converted to primitive ectoderm, Noggin treatment resulted in a dose-dependent reduction of pluripotent markers and an increase in neural and retinal progenitor markers. Interestingly, we also found a substantial number of cells expressing markers for cone photoreceptors in the EPL-driven cultures. These results suggest that first restricting mouse ES cells to a primitive ectoderm lineage creates an environment where Noggin can induce retinal cell marker expression. Conclusions: We are currently determining the underlying molecular mechanism that drives ES cells to retinal progenitors and further, cone photoreceptors Commercial Relationships: Andrea S. Viczian, None; Kimberly A. Wong, None; Michael Trembley, None Support: EY019517 Program Number: 3579 Poster Board Number: A0008 Presentation Time: 3:45 PM–5:30 PM Experimental Analysis of Signaling Pathways Underlying Retinal Cell Specification Using Human Pluripotent Stem Cells Jason S. Meyer1, 2, Akshayalakshmi Sridhar1, Dana Oakes1, Elyse Feder1, Sarah Ohlemacher1. 1Biology, Indiana Univ- Purdue Univ Indianapolis, Indianapolis, IN; 2Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN. Purpose: Human pluripotent stem cells can be differentiated to yield all of the major cell types of the retina, with important implications for studies of retinogenesis as well as degenerative disorders affecting the retina. However, the precise mechanisms underlying the specification of certain retinal cell types over others has been largely ignored. Thus, efforts were undertaken to elucidate signaling pathways responsible for the directed differentiation of major retinal cell types. Methods: Human pluripotent stem cells were differentiated to a highly enriched retinal progenitor cell population following previously established protocols. The ability to influence the differentiation of these retinal progenitor cells was tested through the activation and inhibition of classical signaling pathways, and the effect of these signaling factors on retinal cell fate determination was assessed by immunocytochemistry and qRT-PCR. Differences in cell proliferation and differentiation toward specific retinal cell types was ©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission to reproduce any abstract, contact the ARVO Office at pubs@arvo.org. ARVO 2015 Annual Meeting Abstracts then assessed and quantified, and significant differences between treatment conditions determined. Results: Highly enriched populations of retinal progenitor cells could be derived from human pluripotent stem cells within a total of 30 days of differentiation, expressing progenitor markers including CHX10. By default, these progenitor cells are capable of giving rise to all of the major cell types of the retina, with retinal ganglion cells and photoreceptors the most abundant cell types generated by a total of 70 days of differentiation. Activation and/or inhibition of classical signaling pathways, including Wnt and Hedgehog pathways, demonstrated the ability to alter the differentiation of retinal progenitor cells, biasing their differentiation toward specific retinal neurons. Conclusions: The ability to influence the differentiation of human pluripotent stem cell-derived retinal progenitor cells allows for the unique ability to study critical events of human retinogenesis, yielding enriched populations of retinal neurons. Such an ability also has profound implications for the study of retinal degenerative disorders such as glaucoma or age-related macular degeneration, as enriched populations of specific retinal cell types will likely be required for the development of therapeutic strategies to combat the degenerative processes associated with these diseases. Commercial Relationships: Jason S. Meyer, None; Akshayalakshmi Sridhar, None; Dana Oakes, None; Elyse Feder, None; Sarah Ohlemacher, None Support: NIH R01 EY024984-01, BrightFocus G2012027 Program Number: 3580 Poster Board Number: A0009 Presentation Time: 3:45 PM–5:30 PM Mitochondrial metabolism and structural analysis of Induced Pluripotent Stem Cell (iPSC) derived RPE from Age-Related Macular Degeneration (AMD) Patients Jie Gong1, Mark Fields1, Ernesto F. Moreira1, Hannah Bowrey1, Zsolt Ablonczy1, Baerbel Rohrer1, Craig C. Beeson2, Lucian V. Del Priore1. 1Ophthalmology, MUSC Storm Eye Institute, Charleston, SC; 2College of Pharmacy, MUSC, Charleston, SC. Purpose: Retinal pigment epithelium (RPE) derived from human iPSC may provide a promising source of cells for transplantation. Physiological tests are required to confirm that these cells can function as RPE cells prior to cell transplantation. Herein we analyzed critical RPE functions such as the development of Transepithelial Resistance (TER), polarized secretion of vascular endothelium growth factor (VEGF) and pigment epithelium-derived factor (PEDF) as well as energy metabolism. Methods: Fibroblasts cultured from axillary skin biopsies obtained from a dry AMD patient and an age-matched control were reprogramed into iPSC by using Yamanaka factors (Oct3/4, Sox2, Klf4, c-Myc) delivered using non-integrating Sendai viruses. iPSC cells were subsequently differentiated into RPE cells. iPS (IMR90 Wi-cell Wisconsin USA) was also used as a young control. Pigmented iPSC derived RPE were grown into monolayers on permeable transwells and fed with medium containing 1% serum. Media collected from each chamber were examined for VEGF and PEDF levels using commercially available ELISA kits. TER was measured using an epithelial voltohmmeter. Mitochondrial respiratory function and glycolysis were measured by determining the oxygen consumption rate (OCAR) and extracellular acidification rate (ECAR) of iPS derived RPE using a Seahorse Bioscience XF96 instrument. Results: iPS derived RPE cells from dry AMD and aged control patients formed a hexagonal monolayer exhibiting an RPE phenotype based on pigmentation, the expression of RPE (MITF) and tight junction markers (ZO-1). They form a monolayer with TER of an average of 335+36 Ω*cm2. Polarized VEGF and PEDF secretion was observed [VEGF: basal>apical, (apical 1059+93 pg/ml; basal: 1299+29 pg/ml, p=0.03), PEDF secretion: apical>basal (apical 2185+196 ng/ml basal: 1220+460 ng/ml, p=0.009)]. Mitochondrial respiration and glycolysis revealed an apparent age-dependent decline in metabolic function. However no significance differences between cells from the AMD and the age-matched control sample could be identified. Conclusions: iPS derived RPE cells exhibited physiological properties typically obtained from adult RPE cells. Additional samples are required to confirm this observation and expand it to determine whether there are differences between dry AMD and agematched control samples. Commercial Relationships: Jie Gong, None; Mark Fields, None; Ernesto F. Moreira, None; Hannah Bowrey, None; Zsolt Ablonczy, None; Baerbel Rohrer, None; Craig C. Beeson, None; Lucian V. Del Priore, None Support: Foundation Fighting Blindness, Research to Prevent Blindness and IRB Program Number: 3581 Poster Board Number: A0010 Presentation Time: 3:45 PM–5:30 PM Investigating the Effect of PAX6 Gene Expression on Differentiation of CD133+Human Cord Blood Stem Cells Sahar Balagholi1, 4, Sedigheh Amini Kafiabadi1, Zahra-Soheila Soheili2, Mozhgan Rezaeikanavi4, 3, Shahram Samiei1. 1Iranian Blood Transfusion Organization, Tehran, Iran (the Islamic Republic of); 2National Institute of Genetic Engineering and Biotechnology, Tehran, Iran (the Islamic Republic of); 3Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran (the Islamic Republic of); 4Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran (the Islamic Republic of). Purpose: To assess the influence of PAX6 gene expression on differentiation of CD133+ human cord blood stem cells. Methods: Human cord blood mononuclear cells were isolated by ficoll. CD133+ cells were obtained using the CD133MicroBead Kit in combination with the autoMACS Separator and then cultured in the Stem Span media. HEK293T packaging cells were cotransfected with PLEX-MCS, PsPAX2, and pMD2G via calcium phosphate method. The resultant lentiviral vectors were collected and concentrated by PEG 6000. The appropriate amount of viruses was used for infecting CD133+ cells. The successful transduced cells were selected by puromycin resistance. After two weeks, the expression of rhodopsin, CHX10, Thy1, nestin, and PAX6 proteins were assayed by immunocytochemistry. Results: Two weeks after transfection, CD133+ cells expressed rhodopsin, CHX10, Thy1, nestin, and PAX6 proteins, as compared to the HEK293T cells that expressed rhodopsin and nestin. Conclusions: Transfected CD133+ human cord blood stem cells with PAX6 gene are able to differentiate into progenitor retinal neural- and ganglion-like cells. Commercial Relationships: Sahar Balagholi, None; Sedigheh Amini Kafiabadi, None; Zahra-Soheila Soheili, None; Mozhgan Rezaeikanavi, None; Shahram Samiei, None ©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission to reproduce any abstract, contact the ARVO Office at pubs@arvo.org. ARVO 2015 Annual Meeting Abstracts Program Number: 3582 Poster Board Number: A0011 Presentation Time: 3:45 PM–5:30 PM Modeling retinal degeneration using induced pluripotent stem cells from patients with the common P347L mutation in the RHODOPSIN gene Angelique Terray1, Celine Nanteau1, Amelie Slembrouck1, Jose A. Sahel1, 2, Sacha Reichman1, Isabelle S. Audo1, 2, Olivier Goureau1. 1 Institut de la Vision, INSERM U968 ; Sorbonne Universités, UPMCParis 06 ; CNRS UMR7210, Paris, France; 2Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, Paris, France. Purpose: Inherited retinal degenerations, associated with photoreceptor loss leading to blindness or visual impairment, affect more than one million people throughout the world. The recent discovery of direct reprogramming of somatic cells into induced pluripotent stem (iPS) cells offers the opportunity to model in vitro the effects of mutations on human retinal development. We have focused on the most prevalent form of autosomal dominant retinitis pigmentosa (adRP) in European cohorts, resulting from a mutation (substitution P347L) on the gene coding for the visual pigment RHODOPSIN (RHO). Methods: Reprogramming of fibroblasts is performed by a non integrative approach using Sendai Virus expressing the four reprogramming genes, OCT4, KLF4, SOX2 and C-MYC. The stemness status of the induced pluripotent stem (iPS) cells is assessed phenotypically and by the expression of specific markers both by qPCR and immunohistochemistry. Differentiation of iPS cell lines towards retinal and photoreceptor lineages is performed using a wellestablished protocol (Reichman et al. PNAS 2014; 111:8518). Results: Fibroblasts of three patients from the same family (including one healthy patient) have been successfully reprogrammed into iPS cells. All generated iPS cell lines expressed the specific markers of pluripotency SSEA-4, TRA-1-81, OCT4 and NANOG. Q-PCR analysis confirmed the absence of the Sendai viral reprogramming genes and each iPS cell lines exhibited a normal karyotype. Normal and RHODOPSIN-mutated iPS cell lines were able to generate neural retina (NR)-like structures, containing retinal progenitors within 2 weeks. Culturing these NR-like structures in floating conditions, we have obtained photoreceptor (precursors and matures) from both normal and mutated cells. However after long-term cultures rod and cone cell numbers were significant lower in NR-like structures differentiated from RHODOPSIN-mutated iPS cell lines than in structures differentiated from normal iPS cells. Conclusions: From these results we conclude that mature photoreceptors differentiated from RHODOPSIN-mutated iPS cells degenerate in a RP-specific manner, demonstrating the ability of patient-specific photoreceptors to recapitulate the disease phenotype in vitro. Commercial Relationships: Angelique Terray, None; Celine Nanteau, None; Amelie Slembrouck, None; Jose A. Sahel, None; Sacha Reichman, None; Isabelle S. Audo, None; Olivier Goureau, None Support: Agence Nationale de la Recherche [GPiPS: ANR-2010RFCS005], Région Ile de France (DIM –Biothérapies) and Fondation de France (B. Fouassier). Program Number: 3583 Poster Board Number: A0012 Presentation Time: 3:45 PM–5:30 PM The mechanism of mTOR signaling pathway in the regulation of differentiation of iPS into retinal pigment epithelial cells sijia ding, Chao Jiang, Chen Zhao. The first affiliated hospital with Nanjing Medical University, Nanjing, China. Purpose: To explore the role of mTOR signaling pathway in the regulation of differentiation of iPS into retinal pigment epithelial (RPE) cells. Methods: After embryoid bodies were formed by cultured iPS in suspension condition, they were induced to differentiate into RPE cells. The expressions of RPE specific proteins (RPE65, LRAT, ZO-1) in iPS-RPE cells during differentiation were detected by immunocytochemistry. Q-PCR and Western Blotting were carried out to analyze RPE specific genes, proteins and mTOR activity in iPS-RPE at different time points of differentiation (after one month, two months, three months). Finally, iPS-RPE were treated with mTOR inhibitor rapamycin and RPE specific protein expression was evaluated. Results: The RPE specific proteins (RPE65, LRAT, zo-1) of iPS-RPE cells were observed after one month of differentiation by fluorescence microscope. Compared with the control iPS, Q-PCR results showed that iPS-RPE exhibited significant higher level of RPE specific genes (RPE65, Best1, MerTK, CK18) after three months of differentiation (P <0.01). Western Blotting also showed that the expressions of RPE specific proteins BEST1, catenin and MerTK significantly increased in iPS-RPE cells as differentiation process went on. The activity of the mTOR was inhibited in this process. In addition, rapamycin treated cells exhibited higher expression of catenin, but expression of BEST1, MerTK and CK18 was not changed. Conclusions: We established an efficient method to obtain iPS-RPE cells. And mTOR signaling pathway is gradually suppressed during the process of iPS differentiation into RPE cells in vitro. Commercial Relationships: sijia ding, None; Chao Jiang, None; Chen Zhao, Jiangsu Province‚Äôs Key Provincial Talents Program (No. RC201149) (F) Support: Jiangsu Province’s Key Provincial Talents Program (RC201149 to C.Z.) Program Number: 3584 Poster Board Number: A0013 Presentation Time: 3:45 PM–5:30 PM Retinal Pigment Epithelial Cells Derived from Induced Pluripotent Stem Cells exhibit Cytokine Profiles Similar to Other Human RPE Cell Lines Aya Yanagida, Kaitlen Knight, Jennifer R. Chao. University of Washington, Seattle, WA. Purpose: Age-related macular degeneration is the leading cause of blindness in elderly populations in the developing countries, and currently there are no effective long-term treatments available. In order to better understand AMD disease pathology, there has been significant interest in studying patient-specific iPSC-derived retinal pigmented epithelium (RPE) cells, which are known to play a central role in AMD. To confirm that iPSC-derived RPE cells recapitulate the cell morphology and function of native RPE, we compared them to cultures often used to study AMD, including human fetal RPE, ARPE 19 cells and human embryonic stem cell (hESC) derived RPE. Methods: iPSC- and hESC-derived RPE cells, human fetal RPE (gestational age 18-20 weeks), and ARPE-19 cells were cultured on transwell filter membranes in order to establish cell polarity and monolayers that replicate native RPE. After 4 and 8 weeks, cell cultures were assessed by light, confocal, and transmission electron microscopy (TEM). They were also assessed by transepithelial resistance (TER) measurements in order to detect the formation of tight junctional complexes. Secreted proteins in media (both apical and basal) were analyzed by multiplex protein analysis after 4 and 8 weeks in culture. Results: All cell types expressed RPE markers (CRALBP, ZO-1, and Otx2) by 4 weeks in culture. iPSC-RPE, hESC-RPE, and human fetal RPE, but not ARPE-19 cells, developed tight junctional complexes ©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission to reproduce any abstract, contact the ARVO Office at pubs@arvo.org. ARVO 2015 Annual Meeting Abstracts and apical microvilli, as determined by TEM. In addition, all cell lines except ARPE-19 cells exhibited TER measurements similar to that of native RPE by 8 weeks, indicating the establishment of tight junctions. In a profile of 44 secreted proteins in apical and basal media, iPSC-RPE cells were most similar to hESC-RPE and least similar to ARPE-19 cells. iPSC-RPE were similar to fetal RPE in a polarized (basal > apical) secretion of VEGF-A and apolipoprotein E, two important factors in AMD disease development. Conclusions: Human iPSC-derived RPE cells have similar structure and cytokine profiles compared to native RPE and are the most similar to ES cell derived RPE cells in our study. Our findings provide support for the use of patient–specific iPSC-derived RPE in studying the pathology of retinal degenerative diseases, including AMD. Commercial Relationships: Aya Yanagida, None; Kaitlen Knight, None; Jennifer R. Chao, None Support: Research to Prevent Blindness, NIH K08 EY019714 Program Number: 3585 Poster Board Number: A0014 Presentation Time: 3:45 PM–5:30 PM Use of induced pluripotent stem cell technology to understand photoreceptor cytoskeletal dynamics in retinitis pigmentosa Roly Megaw1, Carla Mellough2, Baljean Dhillon1, Alan Wright3, Majlinda Lako2, Charles ffrench-Constant1. 1Scottish Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom; 2Newcastle University, Newcastle, United Kingdom; 3 University of Edinburgh, Edinburgh, United Kingdom. Purpose: Mutations in RPGR account for 20% of all Retinitis Pigmentosa (RP). RPGR’s function is unknown. It has no cure. Animal rpgr models provide conflicting evidence as to the nature of disease. We set out to establish a novel, human, cell-based model for RPGR disease to test the hypothesis that RPGR mutations lead to retinal degeneration due to a dysregulation of the actin cytoskeleton. Methods: iPSCs were generated from skin biopsies of patients with RPGR mutations (g.ORF15+689_692del4) and unaffected relatives. Our published protocol was modified to produce self-organising 3 dimensional eye cups. RGPR-mutated and control cultures were compared. Results: Mutant and wild-type iPSC lines were generated and characterised. Differentiation of wild type and mutant lines resulted in the generation of optic cups in a self-organising manner. After 100 days in culture, these cups contained organised, mature photoreceptors (PRs), as evidenced by morphology and both RNA and protein expression (recoverin and rhodopsin). RPGR is localised to the PR connecting cilium. Western blot analysis shows expression of a truncated RPGRORF15 protein splice variant in mutated PR cultures compared to controls. PR cultures from RGPR-mutated iPS cells had increased actin polymerisation compared with controls (confocal pixel intensity counts : 59.02 (SD 16.24) vs 23.70 (SD 8.128) p<00081).This finding was confirmed by assessment of F-actin with western blot. Pathways regulating actin turnover were explored. Western blot analysis showed reduction in both Src and ERK phosphorylation in RGPR-mutated PR cultures. An unbiased protein array confirmed this reduction. Several other pathways were also shown to be dysregulated in the RGPR-mutated PR cultures; confirmed by western blots of repeat cultures from varying patient and control cell lines. Conclusions: This study supports the hypothesis that RPGR mutations lead to actin dysregulation. We have identified several pathways which are interupted in RPGR-mutant photoreceptor cultures and could be contributing to disease. This study is the first use (to our knowledge) of human iPSCs with retinitis pigmentosa- causing RPGR mutations to look at pathophysiology of disease. It suggests a role for RPGR in facilitating Rhodopsin transport to PR outer segments by regulating actin turnover. Commercial Relationships: Roly Megaw, None; Carla Mellough, None; Baljean Dhillon, None; Alan Wright, None; Majlinda Lako, None; Charles ffrench-Constant, None Support: Wellcome Trust Grant (100470/Z/12/Z) Program Number: 3586 Poster Board Number: A0015 Presentation Time: 3:45 PM–5:30 PM Immunological properties of human embryonic stem cell-derived retinal pigment epithelial cellsImmunological properties of human embryonic stem cell-derived retinal pigment epithelial cells Qiuhui Liu, Lai Wei, Jing Wang, Shaofen Lin, Xiao Wang, Bing Huang, Lin Lu, Yan Luo. Zhongshan Ophthalmic Center, Guangzhou, China. Purpose: The subretinal transplantation of human embryonic stem cell-derived retinal pigment epithelial cells (hES-RPE) is thought to be one of the most promising therapies for dry AMD and retinitis pigmentosa. Although previous studies indicated that the subretinal space possess immune privilege, the potential immune rejection limits the clinical application of hES-RPE due to the immunological properties of the ES-RPE cells are still unclear. It is necessary to investigate the immunological properties of the ES-RPE cells and develop new strategies to induce immune tolerance of allogeneic transplants. This study was aimed to define the immunological properties of the hES-RPE. Methods: Human embryonic stem cells were induced to RPE cells by adding DKK-1, Noggin and IGF to the medium. Then hES-RPE cells were seed onto matrigel and co-cultured with CD 4+ T cells for 72 hours. The expression of MHC class I and II molecules in the hES-RPE cells before and after co-cultured with CD 4+ T cells were detected by flow cytometry. The production of IL-1 beta, IL-6, TGFbeta 1 and TGF-beta 2 was analyzed by ELISA. Results: The hES-RPE cells expressed specific RPE marker Mitf and RPE65 after 28 days culture, the matured hES-RPE cells had phagocytic ability and could secret PEDF. hES-RPE cells expressed moderate levels of MHC class I molecules and negative for MHC class II molecules. After co-cultured with CD 4+ T cell, the levels of the MHC class I and II molecules slightly increased. Compared with hRPE cells, the level of IL-1 beta and IL-6 produced by ES-RPE cells was less. Cells pretreated with TGF-beta expressed less MHC-II molecules after co-cultured with T cell. Conclusions: ES-RPE cells showed less immunogenicity than hRPE cells; however it still can express MHC class I and II molecules when co-cultured with CD 4+ T cells, indicating it may be rejected after transplantation. TGF-beta might partly suppress the immune rejection. Commercial Relationships: Qiuhui Liu, None; Lai Wei, None; Jing Wang, None; Shaofen Lin, None; Xiao Wang, None; Bing Huang, None; Lin Lu, None; Yan Luo, None Support: National Basic Research Development Program of China (973 program: 2013CB967000) ; the National Natural Science Foundation of China to YAN LUO (81371020) ©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission to reproduce any abstract, contact the ARVO Office at pubs@arvo.org. ARVO 2015 Annual Meeting Abstracts Program Number: 3587 Poster Board Number: A0016 Presentation Time: 3:45 PM–5:30 PM Altered RB and p27 expression patterns in hESC-derived versus fetal retinal cone-precursors Dominic Shayler1, Hardeep P. Singh2, Narine Harutyunyan2, David Cobrinik2, 3. 1Developmental, Stem Cell and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA; 2The Vision Center, Division of Ophthalmology and the Saban Research Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA; 3USC Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA. Purpose: Methods have recently been developed to generate retinal tissue from human embryonic stem cells (hESCs). These hESCderived “retinal spheres” potentially could model aspects of human retinal development and disease more accurately than live animal models. For example, in human but not mouse retinas, maturing cone arrestin (CA)-positive fetal cone precursors have been shown to upregulate RB protein, MDM2 and MYCN, and to downregulate p27.These changes have been implicated in the human cone precursor response to RB loss in retinoblastoma development (Xu et al., 2014, Nature). To investigate whether hESC-derived cone precursors display similar features to fetal cone precursors, as might be needed in a retinoblastoma model, RB, MDM2, MYCN and p27 were examined in spheres of multiple ages. Methods: H9 embryonic stem cells were differentiated into retinal tissue as described (Nakano et al. 2012). Retinal spheres were collected from multiple preparations, frozen sectioned and immunostained. p27, RB and CA expression was observed at days 27, 41, 60/69, 97 and 132, while MDM2 and CA expression was observed at the same ages as well as at day 123. Fetal retina of weeks 15 and 21 served as positive controls. All sections were imaged under confocal microscopy. Results: The earliest CA-positive cells appeared at day 41. Older spheres displayed greater numbers of CA-positive cells with increasing numbers on the apical surface. p27 was downregulated in these cells in fetal retina but not in spheres. RB was expressed at higher levels in cone precursors compared to other retinal neuron cell types in fetal retina, but was expressed at lower levels at all ages in hESC-derived tissue. MDM2 was expressed at higher levels in CApositive cells than other retinal cell types both in retinal spheres and in fetal retina. Conclusions: These data demonstrate that developing CA-positive cells do not share all protein expression patterns with fetal retina. MDM2 displayed similar expression in both systems, but RB was not prominent and p27 remained highly expressed in cone precursors as compared to other retinal cell types in the sphere system. This indicates that there is a difference in expression of proteins that are relevant to retinoblastoma development in the hESC-derived retina model. Commercial Relationships: Dominic Shayler, None; Hardeep P. Singh, None; Narine Harutyunyan, None; David Cobrinik, None Support: The Larry and Celia Moh Foundation, Research to Prevent Blindness, A.B. Reins Foundation Program Number: 3588 Poster Board Number: A0017 Presentation Time: 3:45 PM–5:30 PM Exogenous factors induce rod photoreceptor-specific progenitors from adult mouse retinal stem cells Brian G. Ballios1, Saeed Khalili2, Kenneth Grisé2, Laura Donaldson3, Gilbert Bernier4, Derek van der Kooy2. 1MD/PhD Program, University of Toronto, Toronto, ON, Canada; 2Molecular Genetics, University of Toronto, Toronto, ON, Canada; 3Division of Ophthalmology, McMaster University, Hamilton, ON, Canada; 4 Centre de recherché, Pavillon Marcel-Lamoureux, 4MaisonneuveRosemont Hospital, Montréal, QC, Canada. Purpose: Adult retinal stem cell (RSCs) derived from the ciliary epithelium (CE) of mice can give rise to all retinal cell types. Taurine, retinoic acid and FGF2/heparin (T+RA+FH) added to differentiating clonal RSC colonies increases the number of rods to 90% of all progeny; RSC progeny produce 10% rods when differentiated in 1%FBS+FH (pan-retinal conditions). We hypothesized that T/RA acts on RSC progeny in an instructive, rather than permissive, manner to bias photoreceptor differentiation through the enrichment of rodspecific progenitors. Methods: RSCs were clonally isolated from the CE of 4-6 week old mice. We used limiting dilutions (<1 clone / well) of a fluorescent retroviral construct to label individual progenitor clones in vitro. In addition, single cell sorting isolated non-pigmented and pigmented cells in wells, which were then treated with T/RA for 28 d. Survival, clone size, and phenotype were assessed by immunocytochemistry. Results: Clonal retroviral labeling revealed enrichment in the percentage of rod-only clones between 1%FBS (13%) to T/RA (over 70%), without affecting clone size or overall cell survival. This strongly argues against selective survival of rod progenitors or differential survival of post-mitotic rods within a clone. In 1%FBS, clones derived from single non-pigmented progenitors were distributed between non-rod and mixed clones, with a minority of rod-only clones (100% Rhodopsin-positive; n=4 of 28 clones). Clones derived from pigmented cells in 1%FBS never gave rise to rod-only clones. In T+RA conditions, all clones derived from nonpigmented progenitors (n=34) were rod-only clones, while those derived from pigmented progenitors (n=47 of 48) were almost all norod clones. Of note, one rod-only clone (the largest) was derived from a single pigmented cell in T+RA conditions, suggesting potential neural lineage plasticity in a very early, pigmented progenitor. Survival rates of non-pigmented cell derived clones were similar in T+RA and 1%FBS. Similar experiments using Wnt, BMP4 and TGFβ inhibition increases the number of RSC-derived cones to >60% of all progeny. Conclusions: This study marks an important step in the characterization of a rod-specific progenitor – no markers exist and literature is divided on their existence in vivo. Our study suggests a critical role for exogenous signals instructing early lineage decisions between fate-restricted retinal progenitors. Commercial Relationships: Brian G. Ballios, None; Saeed Khalili, None; Kenneth Grisé, None; Laura Donaldson, None; Gilbert Bernier, None; Derek van der Kooy, None Support: Canadian Institutes of Health Research (CIHR), Foundation Fighting Blindness (FFB) / Krembil Foundation ©2015, Copyright by the Association for Research in Vision and Ophthalmology, Inc., all rights reserved. Go to iovs.org to access the version of record. For permission to reproduce any abstract, contact the ARVO Office at pubs@arvo.org.