Restoring Vision with Photoreceptor Regeneration

Page 1

Zhang C-J and Jin Z-B, J Stem Cell Res Dev 2019, 5: 014 DOI: 10.24966/SRDT-2060/100014

HSOA Journal of Stem Cells Research, Development and Therapy Review Article

Restoring Vision with Photoreceptor Regeneration Chang-Jun Zhang1,2 and Zi-Bing Jin1,2* Laboratory for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research; Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China

1

State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou 325027, China

2

Abstract Neuroretinal diseases are the predominant cause of irreversible blindness worldwide, mainly due to photoreceptor loss. Currently, there are no radical treatments to fully reverse the degeneration or even stop the disease progression. Thus, it is urgent to develop new biological therapeutics for these diseases on the clinical side. Stem cell-based treatments have become a promising therapeutic for Neuroretinal diseases through replacement of damaged cells with photoreceptors and allied cells. To date, great efforts have been made on regenerate the diseased retina based on stem cell technology. In this review, we overview the current status of stem cell-based treatments for photoreceptor regeneration, including the major cell sources derived from different stem cells in pre-clinical or clinical trial stages. Additionally, we discuss herein the major challenges ahead for and potential new strategy toward photoreceptor regeneration. Keywords: Delivery strategy; Neuroretinal diseases; Photoreceptor; Regeneration; Stem cell

Abbreviations QOL: Quality of Life CNS: Central Nerve System RD: Retinal Degeneration RP: Retinitis Pigmentosa AMD: Age Related Macular Degeneration

*Corresponding author: Zi-Bing Jin, Laboratory for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research; Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China, Tel: +86 57788067926; E-mail: jinzb@mail.eye.ac.cn Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014. Received: July 29, 2019; Accepted: August 12, 2019; Published: August 19, 2019 Copyright: © 2019 Zhang C-J and Jin Z-B. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

BM: Bruch’s Membrane; RPE: Retinal Pigment Epithelium; STGD: Stargardt’s Disease RPCs: Retinal Progenitor Cells hRPCs: human-derived RPC MG: Müller Glia NMDA: N-methyl-D-aspartate CE: Ciliary Epithelium IPE: Iris Pigmented Epithelium ESCs: Embryonic Stem Cells iPSCs: Induced Pluripotent Stem Cells hiESCs: human ESCs hiPSCs: human iPSCs MSCs: Mesenchymal Stem Cells ADSCs: Adipose-Derived Stem Cells AESCs: Amniotic Epithelial Stem Cells BMSCs: Bone Marrow Mesenchymal Stem Cells AFMSCs: Amniotic Fluid Mesenchymal Stem Cells 2D: Two-Dimensional 3D: Three-Dimensional LIF: Leukemia Inhibiting Factor BMP: Bone Morphogenetic Protein IGF-1: Insulin-Like Growth Factor-1 SFEB: Serum-Free Floating Culture System ERG: Electroretinogram FACS: Fluorescence Activated Cell Sorting MACS: Magnetic Activated Cell Sorting INL: Inner Retinal Layer ECM: Extracellular Matrix OKT: Optokinetic Testing SD-OCT: Spectral Domain-Optical Coherence Tomography HAMC: Hyaluronan-Methylcellulose HLAs: Human Leukocyte Antigens RA: Retinoic Acid LCA: Leber Congenital Amaurosis GVHD: Graft Versus Host Disease CSNB: Congenital Stationary Night Blindness

Introduction Human eye, the organ of the visual system, is the most important organ of perception and converts visual signals into neural signals in the brain [1,2]. The structure of the human eye is incredibly complex, comprising the cornea, iris and lens from the anterior segment, and the vitreous humor, retina, choroid and sclera from the posterior segment [3,4]. Healthy and asymptomatic eyes are undoubtedly crucial to Quality of Life (QOL). Unfortunately, there are many different diseases and age-related changes that affecting the eye, ranging from the anterior segment to the posterior segment [5,6]. Apart from


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 2 of 22 •

neuroretinal disorders, a good therapeutic effect can be achieved in most eye diseases in the anterior segment with current medical treatments. The retina is a part of the Central Nerve System (CNS) and is primarily composed of seven cell types [7-9]. In the mammalian retina, three vital cell types, including photoreceptors, Retinal Pigment Epithelium (RPE) cells and retinal ganglion cells, lack self-regenerative capacity after injury. Thus, neuroretinal disorders, such as Retinal Degeneration (RD) and glaucoma, can lead to irreversible and incurable visual impairment with the loss of photoreceptors, RPE cells, and/or retinal ganglion cells [10]. Retinal degeneration (RD) encompasses a group of major registered blindness-causing diseases accompanied by the death of photoreceptors and/or RPE cells that result in progressive loss of vision and severely impact the quality of patient’s life [11-14]. RD primarily includes Retinitis Pigmentosa (RP) and macular degeneration, which has a high clinical incidence, and these diseases exhibit different symptoms and result from complex disease-causing factors. RP is an inherited retinal disorder involving more than 90 causal genes, and typical symptoms include progressive night blindness and loss of peripheral visual field [15-17]. RP affects more than 1.5 million individuals worldwide with a prevalence rate of approximately 1 in 4000 [15, 18]. Different from inherited and rod-dominated RP, macular degeneration is an over determined and cone-dominated disease caused by both innate and acquired factors and mainly includes Age-Related Macular Degeneration (AMD) and juvenile macular degeneration [19,20]. The thickened Bruch’s Membrane (BM) is the initial characterization of AMD, and subsequently, the loss of RPE cells and photoreceptors gradually occurs in the late stage [21-26]. It is estimated that the number of AMD cases will reach nearly 196 million worldwide by 2020 and 288 million by 2040, leading to an increased social and economic burden [27-29]. Stargardt’s Disease (STGD) is the highest prevalence of juvenile macular degeneration [30-33]. Both diseases are currently incurable in terms of photoreceptor maintanance and disease reversion. As seen from the foregoing descriptions, traditional therapeutic approaches for neuroretinal diseases can only delay the progression of neuroretinal degeneration and are unable to cure or completely reverse the damage caused by the disease. Unlike the retina in lower vertebrates, the human retina lacks a regenerative capability [34,35]. Stem cells are special cell types characterized by the capacity of unlimited self-renew and the ability of multidirectional differentiation into multiple cell types, including photoreceptors, RPE cells, and/or retinal ganglion cells [36]. In recent years, based on these characteristics of stem cells, stem cell transplantation has been well studied and widely used as a feasible approach for maintaining, enhancing and restoring the function and structure of tissues or organs [37]. In particular, the eye has some natural advantages over other tissues and organs in the field of stem cell transplantation therapy; first, the relatively small size only requires a small volume of cells for transplantation; second, because the eye is easily accessible and observable highly invasive methods for surgery or examination can be avoided; third, the immune privileged sites can help to prevent unwanted inflammatory responses and thus promote the survival of donor cells; fourth, the comparatively independent, separated space can minimize J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

systemic dissemination and maintain the intraocular microenvironment; last, the highly sectionalized structure can permit specific targeted interventions for different ocular tissues [38-41]. During the last two decades, stem cell-based therapy for neuroretinal diseases have become a promising treatment strategy due to the continued advances in generation of appropriate cell sources, development of optimal delivery systems, and exploration of combined cell transplantation.

Stem Cells for Regeneration of the Retina The primary problem facing stem cell transplantation for neuroretinal diseases is identification and characterization of the source of stem cells. Accessible and efficient stem cell sources for neuroretina regeneration are classified into two main categories, endogenous retinal stem cells and exogenous stem cells for retinal regeneration.

Endogenous Retinal Stem Cells The study of restoring vision through retinal transplantation can be traced back to the 1950s, when a donor fetal mouse retina was transplanted into an adult mouse with incomplete integration into the host retina [42]. Subsequently, further studies found that transplantation integration could be increased when the donor or host tissue is at an early stage or the host retina is injured. Retinal Progenitor Cells (RPCs) have been identified as the effective cellular component of fetal or neonatal retinal transplantation [43-45]; transplantation of isolated RPCs promotes integration with the host retina [46,47]. RPCs have a multipotential differentiation capacity, exhibit expression of the neuroectodermal marker nest in and can potentially differentiate into all retinal cells (e.g., Müller glial cells, rod photoreceptors and bipolar neurons) during development [48,49]. In the early stage of mammal retinal development, RPCs, as the predominant cell type, are easy to isolate from several mammalian species, including rodents, pigs, and humans [50-52]. Surprisingly, in addition to cells from fetal or neonatal epiretinal membrane in the adult human retinas, suggesting that the mature retina may harbor RPCs [53,54]. Mouse-, rat-, cat-, and pig-derived RPCs, as well as human-derived RPCs (hRPCs), have been applied to replace damaged cells in corresponding animal disease models with good differentiation and integration [46,55-59]. However, the low rate of RPC cell proliferation and ethical issues regarding clinical application of RPCs harvested from fetal or neonatal eyes limit their transplantation into human patients. For the treatment of retinal degenerative diseases, in addition to attempts to replace damaged cells through stem cell transplantation, the endogenous ocular stem cell populations can be reactivated to generate new retinal photoreceptors [60-62]. The well-known endogenous retinal stem cells Müller Glia (MG) are derived from the retinal neuroepithelium and are the major glial unable to the retina [63,64]. MG span the entire length of this structure and function to maintain retinal homeostasis and integrity [65]. Unlike most cell types, the differentiated retinal neurons are incompetent to divide and regenerate once damaged or lost. Surprisingly, MG has a prominent ability to regenerate retinal tissue; however, the capacity for MG regeneration varies greatly among different species. In some non-mammalian species, such as chick, fish and birds, MG have an ability to regenerate neurons [66-68]. In zebrafish, MG generate various retinal neurons and restore visual function through a reprogramming event. In contrast to zebrafish, mammalian MG encounter reactive gliosis and Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 3 of 22 •

exhibit hypertrophy after retinal impairment but fail to regenerate novel neurons to replace lost cells under natural circumstances [69]. Specially, mammalian MG can also proliferate and regenerate the adult mammalian retina in response to N-methyl-D-aspartate (NMDA) neurotoxicity in the adult rat retina [70,71]. Further studies showed that the mammalian retina is also capable of regenerating inner retinal neurons after treatment with specific growth factors in mouse retina with intraocular NMDA [72]. Likewise, following NMDA injection in rats, MG started to proliferate mediated by cyclin D1- and D3- related pathways and differentiated exclusively into rhodopsin-positive cells surrounded by synaptophysin, showing the potential for synapse formation [73]. In addition to these neurotoxic substances, axotomy and laser injury can also result in proliferation and differentiation of MG into retinal neurons in vivo [74,75]. Furthermore, molecular fingerprinting showed that MG express genes associated with retinal stem cells in the mouse retina [76]. Although the differentiation potential of endogenous human MG has not yet been shown, in vitro, MG isolated from adult human retinas was shown to possess an ability to differentiate into rod photoreceptors for the first time [77]. Moreover, human MG-derived photoreceptors and MG-derived retinal ganglion cells were transplanted into deficient rodent retinas, resulting in the functional restoration of rod or retinal ganglion cells [78-80]. Although some previous studies have shown that the newly generated neurons are functional and can partially restore visual function, neurogenesis is still limited, evidenced by the low number of regenerated cells. Thus, novel strategies are required to maintain adequate numbers of MG and promote their effective trans-differentiation to regenerate newly formed retinal neurons in vivo. As with MG, many previous studies showed that some other ocular stem cells, including RPE [81,82], the Ciliary Epithelium (CE) [83,84], and Iris Pigmented Epithelium (IPE) [85-87], also retain some restricted regenerative capacities. However, the efficiency of reprogramming and the potential of produced new cells derived from these endogenous stem cells are so low that they do not achieve extensive replacement of mature mammalian eyes after injury or disease. Based on the advancement of in vitro culture techniques, they may also be promising as a source of donor cells for cell replacement therapy. Moreover, the development of new techniques to induce reactivation and differentiation of endogenous stem cells is another breakthrough in the treatment of neuroretinal diseases.

Exogenous Stem Cells Exogenous stem cells for retinal regeneration are primarily non-ocular stem cells. The non-ocular stem cells that can generate retinal cells are mainly divided into three types, Embryonic Stem Cells (ESCs) [88], induced Pluripotent Stem Cells (iPSCs) induced from mature somatic cells [89], and Mesenchymal Stem Cells (MSCs) [90]. MSCs can be obtained from various sources, including Adipose-Derived Stem Cells (ADSCs) [91], Amniotic Epithelial Stem Cells (AESCs) [92], Bone Marrow Mesenchymal Stem Cells (BMSCs) [93], and Amniotic Fluid Mesenchymal Stem Cells (AFMSCs) [94]. ESCs and iPSCs are currently widely used and have been selected as prospective therapeutic strategies for neuroretinal diseases; thus, they are our next narrative focus. J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

Differentiation of ESCs and iPSCs into Photoreceptors and RPE Cells The first pluripotential cells from mouse embryos were established by Martin, Evans and Kaufman in 1981 [95]. Subsequently, Tomson and colleagues isolated ESCs from primate and human embryos in 1998 [88]. ESCs and iPSCs are capable of differentiation into various retinal cell types using Two-Dimensional (2D) or Three-Dimensional (3D) culture systems, including photoreceptors [96,97], RPE cells [98,99], and retinal ganglion cells [97,100].

Deriving Photoreceptors from ESCs and iPSCs Direct differentiation of ESCs into neural and retinal cell types can be guided by manipulation of signaling pathways in vivo, sequentially followed by the withdrawal of Wnt signaling, Leukemia Inhibiting Factor (LIF), and Bone Morphogenetic Protein (BMP) and the activation of Insulin-Like Growth Factor 1 (IGF-1) and Activin/Nodal signaling pathways [96,101-109]. Mouse ESC-derived neuroretinal cells have been induced in vivo by using certain factors for neural and retinal induction [106]. Mouse ESCs successfully generated retinal progenitors in the presence of FBS at reasonable efficiency (26%) after treatment in A Serum-Free Floating Culture System (SFEB) with addition of Dkk1 (a Wnt antagonist), Lefty A (a nodal antagonist), serum and activin, successively. However, only in co-culture with embryonic retinal cells can these retinal progenitors efficiently differentiate into photoreceptor precursors [106,110]. Subsequently, human ESCs were reported to produce neural progenitors in vitro, but their differentiation into retinal cells needs to be performed via intraocular transplantation into adult rats [111]. Similarly, human ESC-derived retinal progenitors were induced under the addition of dkk1, noggin and IGF-1 in Matrigel and continued to generate photoreceptors in co-culture with adult mouse retinal cells [112]. A groundbreaking work showed that mouse, monkey and human ESC-derived photoreceptor precursors was realized under optimized culture conditions in vitro, with elimination of the need for animal-derived substances or retinal tissue [113]. Ethical issues and immune rejection are major constraints in clinical application of hESCs [114]. In 2006 and 2007, mouse and human somatic cells were induced for the first time to form the pluripotent stem cells via reprogramming using the “Yamanaka factors”, Oct3/4, c-Myc, Sox2, and Klf4 [89,117]. These iPSCs pose identical self-renewal and multi-differentiation properties as ESCs and serve as a prospective source of donor cells to eliminate or reduce the immune rejection by using the iPSC-derived retinal cells from the patients. Mouse and human iPSCs can differentiate into photoreceptors with defined factors in vitro through the stepwise differentiation process used in differentiation of ESCs [105]. In summary, the 2D-based protocols used in differentiation of ESCs or iPSCs toward photoreceptors all proceed through an analogous regulatory pathway, including suppression of endogenous BMP/TGFβ, Activin/Nodal and Wnt signaling pathways with the addition of IGF-1 or Insulin and combination with various regulatory factors (such as FGF1/FGF2/T3/ COCO), retinoic acid (RA) and taurine [105,107,113,116-123]. To change the low differentiation efficacy observed in 2D culture systems and establish robust protocols that provide a sufficient number of donor cells, mouse and human ESC-derived optic cups were first generated in the Sasai lab using a 3D culture system; these optic Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 4 of 22 •

cups are a significantly multilayered retinal tissue composed of major neural retinal components, generally referred to as retinal organoids [124,125]. Moreover, hiPSC-derived optic vesicle-like structures or retinal organoids grown using 3D culture systems have also been established [126-125]. On the basis of 3D retinal organoid technology, many researchers have cultivated and produced a large number of ESC/iPSC-derived photoreceptor cells for transplantation research [129-146]. In this review, we will not go into too much detail. In addition to generating transplantable cells or tissue, 3D retinal organoid technology also holds great potential for in vitro disease model, large-scale drug screening and testing, and cell development exploration [147,148]. Patient iPSC-derived optic cups were used to investigate the disease mechanism of hereditary blindness with the common CEP290 mutation [146]. Most recently, our lab generated iPSC-derived retinal organoids from RP patient with RPGR gene mutations, which can monitor photoreceptor degeneration similar to retinal morphology of the RP patient [149]. Interestingly, these defects were partially rescued byCRISPR-Cas9-mediated correction of RPGR mutation.

Derivation of Photoreceptor-Supportive RPE Cells from Pluripotent Stem Cells Considerable progress has been made in developing efficient protocols to direct differentiation of both ESCs and iPSCs into RPE cells, and thus, a plentiful and controllable source of RPE cells can be serviceable for cell transplantation therapy. An early study showed that mouse ESCs were efficiently induced to differentiate into melanocytes in vitro [150]. Subsequent studies demonstrated that RPE cells could also be derived from mouse and primate ESCs [151,152]. The first hESC-derived RPE was generated using the original protocol, a differentiation system without coculture using certain cells or factors developed by Klimanskaya’s group [153]. Transcriptomics analysis of hESC-derived RPE, mature RPE and human fetal RPE confirmed that they are more similar to native RPE tissue than the existing human RPE cell lines. Vugler and colleagues found that hESC-derived RPE cells express several markers associated with the development and maturity of RPE cells, including Pax6, OTX1/2, RPE65 and PMEL17 [154]. Although ESC-derived RPE cells have features very similar to those of typical RPE cells in morphology and function, the differences of in vitro cell maturity require further analysis to obtain optimal transplant cell populations for transplantation. Theoretically, an unlimited source of RPE cells can be harvested utilizing this culture system. Since then, different labs have optimized the protocol to derive pure and safe RPE cells from hESCs and iPSCs [116,155-167]. As described earlier, iPSCs possess regenerative capability equivalent to those of hESCs and thus are also used as another pluripotent cell source for cell and tissue regeneration [89,115,167]. The process of iPSC-derived RPE cell differentiation can be greatly promoted directed by inhibiting the signaling pathways of Wnt and Nodal in RPE cells with the addition of factors such as Dkk1 and Lefty-A [172,173]. Morphologically and functionally, iPSC-derived RPE cells have the similarity with native RPE and express RPE-specific markers. In view of the fact iPSCs can be generated from any differentiated somatic cells by induction with “Yamanaka factors” [89,174], patient-specific iPSCs afford a potential cell replacement therapy that theoretically may circumvent immune rejection [175-177]. J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

In summary, iPSC-derived RPE cells are a promising cell source to replace defective RPE cells in retinal degeneration.

Transplantation of Stem Cell-Derived Photoreceptors, RPE for Retinal Repair Above, we have introduced stem cell sources of photoreceptors from stem cells above that are considered efficient cell sources for photoreceptor transplantation target to neuroretinal degenerative diseases. From the first attempts at transplantation of human ESC-derived neural progenitors into immunodeficient rats intravitreally and subretinally, which showed an insufficient sources for photoreceptor transplantation [178], to the preliminary successful research showing that human ESC/iPSC-derived neural progenitors could effectively protect vision in RCS rats (an AMD-like rat model) [179,180], the cumulative findings during the past few decades have encouraged researchers to further explore transplantation of retinal specific cell types.

Transplantation of Stem Cell-Derived RPE Cells: Preclinical Studies and Clinical Trials Previous studies of allogeneic and xenogeneic replacement therapy using autologous or mature RPE cells showed an unstable phenotype and a limited source of RPE cells, which provided a space for development of cell replacement therapies using stem cell-derived RPE [153,181-197]. Stem cell-derived RPE cells have been the focus of replacement therapy for AMD and STGD, which are assigned to macular disorders characterized by early loss of the RPE cells that maintain the survival of photoreceptors. ESC-derived RPE cells were first transplanted as a monolayer to treat RPE dysfunction in RCS rats using primate ESCs and enhanced the survival of the host photoreceptors [198]. Subsequently, hESC-derived RPE cells were also utilized for subretinal transplantation in RCS rats and Elovl4 mutant mice (a STGD-like mouse model) and safely prompted preservation of visual function and production of more photoreceptors [154,155,199-201]. Likewise, hiPSC-derived RPE cells were also able to rescue vision in animal models of retinal degeneration [185,202-205]. These preclinical studies have set an encouraging and solid foundation for clinical trials of human ESC/ iPSC-derived RPE transplantation. In 2011, the U.S. Food and Drug Administration approved phase I/II clinical trials for ESC-derived RPE transplantation in the treatment of macular degeneration [206]. According to the current registered clinical trials using stem cell-based therapies for STGD or AMD (https://clinicaltrials.gov/), phase I and II clinical trials are in progress for RPE replacement using hESC/iPSC-derived RPE cells in single-cell suspensions or in monolayers seeded on scaffolds. The preclinical study and clinical trial of hESC-derived RPE cell suspension (named MA09-RPE, forming a master cell bank of hESCs) for treatment of non-neovascular AMD and advanced STGD was first launched in America and primarily demonstrated the safety of subretinal transplantation of hESC-derived RPE cell suspension [206208]. In addition, the potential efficacy has also been confirmed by clinical trials (NCT02463344, NCT02563782) for non-neovascular AMD using a cell suspension with immunosuppressive therapy as resistance to rejection. Moreover, the same MA09-RPE cell suspension was used to treat non-neovascular AMD and STGD in a phase I/IIa Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 5 of 22 •

clinical trial in South Korea without abnormal proliferation and tumor formation [209]. In addition to the above therapeutic strategies using a stem cell-derived RPE cell suspension, an alternative optimized approach is to grow RPE cells on a bioengineered scaffold to form an RPE monolayer before transplantation, which can improve resistance to adverse events and produce polarized RPE cells in a highly functionalized cell monolayer [210-214]. As with CPCB-RPE1, hESC-derived RPE cells are seeded on a biosynthetic scaffold designed to mimic BM, produced as described previously [215,226]. Several preclinical studies were performed to evaluate the transplantation effects of CPCB-RPE1 into the RCS rats and Yucatan mini pigs, and the results indicated that CPCB-RPE1 implants survived in an intact monolayer without development of intraocular or systemic tumors [215-217]. Then, the safety and efficacy of CPCB-RPE1 transplantation was confirmed in a phase I/II a clinical trial for non-neovascular AMD in America, and visual improvement in the short term was observed in some patients. In England, an RPE patch named PF-05206388, composed of hESC-derived RPE monolayer on a coated and synthetic basement membrane, was implanted into the subretinal space of severe neovascular AMD patients, resulting in an increase in visual acuity over 12 months. Unwilling to lag behind, China has initiated two phase 0/I clinical trials for treatment of AMD conducted by Beijing Tongren hospital (NCT02755428) and Southwest hospital (NCT02749734). The phase I clinical trials for neovascular AMD (NCT02749734) using a hESC-derived RPE cell suspension showed a limited functional improvement in vision, albeit to different degrees between patients [205]. Implantation studies of hiPSC-derived RPE cells have lagged behind those of hESC-derived RPE cells. However, allograft transplantation of hESC-derived RPE requires lifelong immunosuppressive therapy, and in contrast, iPSCs do not provoke an immune reaction [218,219]. Due to allowance for autologous transplantation, reduction of ethical issues and potentially unlimited cell sources, hiPSC-derived RPE cells have become widely used. Moreover, particularly, patient iPSC-derived RPE is considered the best in vitro disease modeling candidate to visually and accessibly monitor progression of human disease progression [197,220-222]. A preclinical study in Rpe65rd12/Rpe65rd12 mice showed that implantation of hiPSC-derived RPE cell suspension into this mouse model safely presented colocalization with the host native RPE cells and restored modest vision detected by Electroretinogram (ERG) [223]. Another preclinical study in RCS rats demonstrate that hiPSC-derived RPE cells have the ability to phagocytose photoreceptor material and maintain long-term visual function after transplantation of a cell suspension [185]. The other preclinical studies in RCS rats reported that transplantation of hiPSC-derived RPE cell suspension exhibited protective effects and restored visual function. [197,202,214,224]. To optimize the beneficial effects to host retinal function, an intact donor hiPSC-derived RPE monolayer grown on biomaterial scaffolds was implanted, similar to hESC-derived RPE. This novel approach for hESC-derived RPE cells as apolarized monolayer on a thick synthetic scaffold was first transplanted into RCS rats [225], and subsequently, hESC-derived RPE cell patch or sheets on biodegradable scaffolds were used for AMD treatment in rodents and pigs and provided positive results for clinical trials [226-228]. J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

The progress in iPSC replacement therapy is cruising to the clinic. In 2013, a clinical trial authority for hiPSC-derived RPE into patient treatment was approved in Japan [229]. In 2014, Takahashi’s group from the RIKEN Institute safely performed the first transplantation of autologous iPSC-derived RPE sheets for a female neovascular AMD patient in clinical trial. However, in 2015, RIKEN suspended the clinical trial for another patient due to genomic variations in iPSCs [230]. The landmark research results were finally published in 2017 and showed that the best visual acuity correction had not been improved or worsened 1 year after transplantation [137]. Currently, a novel clinical trial for autologous hiPSC-derived RPE transplantation in AMD is ongoing by Moor fields eye hospital (NCT02464956).

Transplantation of Stem Cell-Derived Photoreceptors The initial studies of photoreceptor replacement focused on photoreceptors derived from donor tissues [231-233], which are known as a very limited cell sources for clinical application. It was demonstrated that the development stage of donor cells influenced the grafting outcomes, and post-mitotic photoreceptor precursors isolated from P4 mouse retina were an optimal donor cell [231,234]. Then, transplantation of these photoreceptor cells successfully restored functional rod-mediated vision in Gnat1−/− mice [233], a model of congenital stationary night blindness with dysfunctional rods [235]. The fact that less mature photoreceptor precursors derived from mouse ESCs helped to improve transplantation success was again confirmed [132,236]. Numerous transplantation studies [129,132,135,139,224,241-239] have been subsequently performed with retinal cells differentiated from mouse ESCs and iPSCs generated using the differentiation protocol reported by Eiraku and Tucker [124,224] (Table 1). To obtain abundant photoreceptors derived from mouse ESC and iPSC and minimize potential tumor genesis from undifferentiated cells, cell-sorting strategies have been used to purify miscellaneous cell populations prior to transplantation [224,237]. These cell-sorting strategies via Fluorescence Activated Cell Sorting (FACS) and Magnetic Activated Cell Sorting (MACS) were established based on the expression of photoreceptor-specific fluorescent reporters or cell surface markers [233, 240-242]. They have been successfully used to purify mouse ESC- and iPSC-derived photoreceptors before transplantation [132,135,139,236,237]. These studies from different groups demonstrated that mouse ESC- and iPSC-derived photoreceptors have the competence to partially recover visual function in various mouse models associated with RD. In 1997, for the first time, a human photoreceptor obtained from adult human cadaveric eyes was successfully implanted into two RP patients but was unable to improve vision [243]. And transplantation of human fetal retinal sheets into RP patients showed that they are capable of surviving and improving vision partially with no rejection of the allogeneic transplant [244-246]. However, ethical issues and the limited source of fetal tissue-derived retinal sheets are bound to make it impossible to be used on a large scale. Early, neural progenitors deriving from differentiation of hESCs were implanted into immune-suppressed rats and non-immunosuppressed mouse retinas, and the results indicated that they could survive for a long time without tumor formation but differentiated into photoreceptor-like cells with a low-level of efficiency [103,247]. Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 6 of 22 •

Subsequently, following transplantation into RCS rats, neural progenitors deriving human ESC and iPSC continued to present uncommitted progenitors with Nestin expression but no expression of specific retinal markers [248,249]. However, vision loss was significantly retarded in RCS rats although not completely halted, which may be mediated by phagotrophic capacity toward photoreceptor outer segments or secretion of neurotrophic factors from the injected neural progenitors [250]. In the earliest transplantation studies of human ESC- derived retinal cells (80%), they were transplanted into the subretinal space of Crx-/- mice (a mouse model of Leber’s Congenital Amaurosis) [251] and differentiated into functional photoreceptors and restored light responses in the mouse model 2–3 weeks after injection [107]. Moreover, human iPSC-derived photoreceptors purified via FACS in combination with a photoreceptor-specific GFP marker could integrate into the host ONL of adult wild-type mice, similar to normal mouse photoreceptors and human ESC-derived rod photoreceptors [107]. Similarly, human ESC- and iPSC-derived photoreceptor progenitors, following FACS or MACS sorting, differentiated into photoreceptors after transplantation into rd1 mice, and integrated into the host retinal neural circuit, restoring partial visual function [240]. In a recent study, human iPSC-derived mature photoreceptors brought mild recovery of host light perception when transplanted into rhodopsin mutant nude rats and primate models of retinal degeneration [252]. For the rod-dominated mouse retina, transplantation of donor rod photoreceptors into murine models of retinal disease improved vision to different degrees [233,234,240,253]. Theoretically, for cone-dominated human retinas, cone photoreceptor replacement should be the optimal strategy for treatment of human retinal degeneration. Several reports of cone transplantation showed a detectable restoration of visual function using a variety of cone and cone-like donor cells in degenerative hosts [238,254-256], which resulted from donor cone integration into degenerative retina. Meanwhile, a significant proportion of material transfer between donor cells and the host retina was observed in non-degenerative hosts [238, 254-261]. These clinical trials mentioned above for treating RD using stem cell-derived RPE have aroused a strong interest in promoting stem cell-derived photoreceptors toward clinical applications. One landmark step is that human ESC lines for therapeutic purposes have been derived from Good Manufacturing Practices (GMP)-compliant xeno-free and feeder-free protocols [262]. Moreover, fullly GMP-compliant human iPSC lines have been generated from adult fibroblasts with different inherited retinal diseases in patients [263]. Then, human ESC- and iPSC-derived photoreceptors were generated using feeder-free and xeno-free GMP conditions [125,127, 263-265].

Strategies for Delivering Stem Cell-Derived Photoreceptors, RPE Cells Stem cell-derived retinal cells are often injected into the eye via direct injection intravitreally or subretinally. Intravitreal injection is easier and less invasive and provides a more access to the Inner Retinal Layer (INL). However, this delivery route is associated with a risk of short-term complications, including retinal detachment, endophthalmitis, and hemorrhage [266,267]. Subretinal injection offers a direct and effective route to deliver donor cells with more precise localization via a minimally invasive J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

injection [206,268]. To avoid the complications and improve the injection effectiveness, subretinal delivery requires skill and stability until mastered. The routes of subretinal injection can be classified into three types: a trans-corneal route passing the lens and vitreous humor [269-272], a trans-scleral route entering the pars plana at the limbus or ora serrata [273-276]; and a transscleral route through the choroid and BM without penetrating the retina [277-279]. The choice of different routes depends on the anatomy of the eyeball according to the species and age. Bleb formation in the subretinal space, come down to retinal detachment that creates a space between the outer segment of the photoreceptor and the RPE, is defined as a successful sign of injection.

Delivering Stem Cell-Derived RPE Stem cell-derived RPE is implanted into the subretinal space as an isolated cell suspension or monolayer on a scaffold [285,286]. After subretinal injection, the scattered RPE cell suspension forms aggregates without an intact monolayer, initiates an immune response [282], and causes severe complications, such as proliferative vitreoretinopathy [215,283]. The use of biocompatible scaffolds [283-287] could avoid these issues and enables proper subretinal delivery of stem cell-derived RPE cells as a monolayer structure for transplantation [288]. Thus, monolayer RPE sheets can be constructed with naturally occurring polymers or synthetic scaffolds. Natural materials are reported to potentially cause a series of issues, such as risk of infection, difficulties controlling the consistency of the material, and mechanical instability [289,290]. Synthetic scaffolds are widely used due to their biocompatibility, ability to imitate the native Extracellular Matrix (ECM), and biodegradability [291-293]. Particularly, parylene, an undegradable synthetic material, is widely used in RPE sheet construction to aid in an intact formation of polarized monolayer without disruption of host RPE structure [210,216,294], which can promote donor RPE cells to survive and integrate into the host RPE [216]. As mentioned above, CPCB-RPE1, an hESC-derived RPE monolayer seeded on an ultrathin polymeric parylene-C scaffold, has been tested in a clinical trial [215,216]. Studies of other synthetic materials (i.e., PLGA, PET, and PCL) are also ongoing [225,295-297]. In addition, RPE sheet can be generated on a collagen coating without an additional scaffold and then released from the digested collagen coating [226]. Furthermore, a human iPSC-derived RPE sheet generated using this strategy has been transplanted onto one AMD patient [137]. Scaffold-based subretinal delivery requires a larger retinotomy into the subretinal space compared with dispersed cell delivery, and some efforts have to be made for more subtle manipulation and smaller microfabrication.

Delivering Stem Cell-Derived Photoreceptors The challenges of photoreceptor transplantation primarily involve how to improve the survival rate of transplanted photoreceptors and promote integration of transplanted cells with the host retina to restore vision function. The key step in photoreceptor transplantation is to guarantee effective delivery of donor photoreceptors into the host subretinal space. The strategies for donor photoreceptor delivery can primarily be divided into three approaches: injection of photoreceptor cell suspensions, transplantation of intact retinal or photoreceptor sheets and transplantation of photoreceptors seeded onto engineered scaffolds. Although these approaches have been applied for treatment Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 7 of 22 •

of photoreceptor degeneration and achieved an encouraging result, there is no single approach that perfectly solves all the problems [236,283,298-303]. Before transplantation studies toward stem cell-derived photoreceptors, transplantation of primary photoreceptors achieved considerable progress using a single-cell suspension approach [233,253,255,304]. Then, cell suspension transplantation of ESC- and iPSC-derived photoreceptors into mouse models of RD improved vision function as measured by ERG [107,109,224,305]. Notwithstanding the tactic of cell suspension transplantation has some crucial advantages, including precise control of the number of transplanted cells and minimally invasive delivery via subretinal injection [107,109,224,305], it also has some unexpected disadvantages, including high incidence of cell death of transplanted cells and low rates of cell survival and integration [129,306-309]. Transplantation of retinal sheets has been studied as an alternative tactics for photoreceptor replacement in RD. Many previous studies of photoreceptor replacement focused on the usage of intact retinal or photoreceptor sheets [232,243,246,310-312], but these individuals with these transplants presented large variations in visual function recovery. Similarly, mouse ESC- and iPSC-derived retinal sheets differentiated into mature photoreceptors with OSs and integrated into the host retina, restoring partial vision after transplantation into rd1 mice [129,239]; but hESC-derived retinal sheets were successfully transplanted into monkey and rat models of retinal degeneration, and then matured into structured grafts without positive functional changes in monkeys and resulted in visual function recovery as measured by Optokinetic Testing (OKT) in rats [313,314]. In addition to variations in visual function recovery, retinal sheet transplantation relatively highly invasive for the patient and often leads to an immune response to the transplants [315,316] compared with single-cell suspension. To further improve photoreceptor transplantation, biomaterial scaffolds designed for photoreceptor delivery have been developed. They can enhance cell survival and promote cell integration into the host retina. Many biomaterial scaffolds have been constructed using multiple polymers alone or jointly, such as PHBV8, PLLA, PLGA, PMMA, PGS, PCL, HA, PDMS, and MC, and have been applied in transplantation of mouse or porcine RPCs [288,306-308,317-322]. Biodegradable scaffolds with high permeability have an insurmountable advantage in promoting photoreceptor survival with nutrient and oxygen diffusion [225,304,318].

Future Directions and Challenges Ahead for Photoreceptor Regeneration As described above, stem cell-based therapies for neuroretinal diseases have achieved unprecedented progress in the last decades. With the progress in cell transplantation therapies, perhaps in the next 10 years, some neuroretinal diseases will be completely cured. Currently, transplantation studies of different stem cell-derived retinal cells are at different stages of development. Stem cell-derived RPE transplantation has been pushed into clinical trials and produced positive preclinical data. Due to the complex circuitry of photoreceptors within the inner retina or optic nerve, stem cell-derived photoreceptor replacement still lies at the preclinical study level with positive results. In short, different grand challenges exist for the two cell transplantation therapies and remain to be overcome in the future.

Stem Cell-Derived RPE Transplantation ESC- and iPSC-derived RPE transplantation has been performed in numerous preclinical studies [154,155,185,197-199,215217,225,226,228,324,325] and clinical trials [137,206-208,326,327]. After human ESC-derived RPE transplantation into AMD or STGD patients, no tumor formation or immunological rejection was observed, confirming the safety [326,328]. However, no significant visual improvement was observed despite visible pigmentation and thickening of the RPE layer in the host retina detected by noninvasive fundus photography and Spectral Domain Optical Coherence Tomography (SD-OCT). Therefore, it is especially urgent to explore new strategies to promote curative effects. The viability of RPE cells was significantly promoted by culture in Hyaluronan-Methylcellulose (HAMC) hydrogel combined with IGF-1 factor, which was illuminated by a study in which dual-use biocompatible materials can be used as drug delivery platforms or to encapsulate cells [329,330], indicating that strategies for co-delivery of growth factors and retinal cells to improve the curative effects of stem cell-derived RPE transplantation are promising. Future research needs to focus on this interesting point.

Furthermore, recent studies showed that 3D micro structured scaffolds promote efficient polarization of human iPSC-derived photoreceptors and basal axon extensions [323]. And two-photon polymerized polycaprolactone successfully supported human iPSC-derived retinal progenitor cells for implantation into a porcine model of retinitis pigmentosa [297]. These pioneering studies on the delivery of stem cell-derived photoreceptors combined with biomaterial scaffolds pave the way for future photoreceptor transplantation therapies.

Additionally, human iPSC-derived RPE has been reported [331] to exhibit differential expression of ES-associated miRNAs compared with human fetal RPE, and thus underscoring the importance of assessing human iPSC-derived RPE completely before transplantation. Moreover, a clinical trial was suspended due to genomic variations found in the transplanted human allogeneic iPSCs [230], which may have a potential risk of tumorigenicity. To improve the reliability of stem cell-based treatment, donor iPSCs or iPSC-derived RPE cells must be subjected to extensive testing. Donor iPSCs induced from autologous somatic cells will be a good choice for stem cell-based treatment. For future allogeneic trials, a GMP-compliant bank of iPSCs, similar to that of ESCs, should be gradually established in all qualified medical institutions [230]. And these cells should be matched with patient Human Leukocyte Antigens (HLAs), thereby reducing the risk of immunological rejection [332-335].

Taken together, these strategies for delivering stem cell-derived retinal cells in combination with biodegradable scaffolds are very promising advanced techniques to treat and even cure neuroretinal diseases in the future.

In addition to the above burning problems, standardized cell differentiation, high-efficiency cell delivery, preferable safety evaluation, most suitable phase for transplantation therapies and therapeutic analysis are all unremitting pursuits for future clinical trials.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 8 of 22 •

Stem Cell-Derived Photoreceptor Transplantation A number of transplantation studies using stem cell-derived photoreceptors have been carried out in many photoreceptor-deficient animal models (Table 1). Although the integration of transplanted photoreceptors into host retinas has been reported in some previous studies [107,237,240,336], vision improvement was not significantly observed due to insufficient integration of cells. Therefore, the first issue is how to obtained sufficient donor cells in an efficient and reliable manner. To overcome this issue, on the basis of fully GMP-compliant generation of human ESC- and iPSC-derived photoreceptors [125,127,263,265,337], an efficient and reliable cell sorting strategy could be added to the GMP guidelines. Certain cell-specific surface antigens (CD73 or C-Kit) have been used as markers for sorting photoreceptors from human fetal retinas or stem cell-derived organoids via MACS or FACS [205,338-340]. In the future, achievement of a GMP-compliant photoreceptor sorting strategy will lay the foundation for acquiring appropriate donor cells. The second outstanding issue is how to interpret and identify material transfer or cell integration between donor cells and host retina after photoreceptor transplantation into animal models. Before material transfer (RNA and/or protein) was found in donor cells and host photoreceptors [257,258,341], the therapeutic efficiency of photoreceptor replacement was attributed to its integration into the host retina and differentiation of rods [107,109,233,234]. Human ESC- and iPSC-derived photoreceptors have been confirmed to integrate and form synaptic connections with host cells that result in vision improvement [129,239,305], and material transfer and cell integration could occur at the same time at different ratios [339,342-345]. However, more work needs to be performed to illuminate the cellular mechanisms of this phenomenon. The third general issue is how to improve the effectiveness of treatments based on human iPSC-derived photoreceptor transplantation. As is known, autologous transplantation of photoreceptors derived from the patient’s own iPSCs will evade immunological rejection. However, iPSCs from patients still carry the disease-causing gene mutations, leading to potential risk of pathopoiesis after transplantation. CRISPR-Cas9 gene editing technology allows modification of the gene mutations in patient iPSCs during in vitro culture [346]. Our group has corrected an RPGR mutation in patient iPSCs using CRISPR/Cas9 gene editing technology in vitro, which rescued photoreceptor structure and electrophysiological property in retinal organoids derived from these iPSCs [149]. Therefore, human iPSC-derived photoreceptors with CRISPR-Cas9-mediated correction will be a promising strategy for treatment of inherited retinal disorders. How to definitively solve cone transplantation is also a worthy concern. Previously, the vast majority of studies primarily focused on rod transplantation and demonstrated that transplanted donor rod photoreceptors possess the ability to rescue vision impairment in rodent models of retinal diseases [109,190,231,234,236,240,256,303,339,342345,347]. Notwithstanding, cone transplantation has also been reported recently [254-256], and these transplanted donor cones were characterized by rod-like morphological features, which were deemed J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

to result from cell integration and/or material transfer determined by the different host microenvironments [238], but detailed mechanisms need to be further explored to improve the efficiency of cone transplantation. Actually, the influencing factors in photoreceptor transplantation are more than those mentioned above. Suitable preclinical animal models, immune rejection, tumor formation, cell survival, and delivery systems should all be further optimized carefully and continuously before clinical trials.

The Expected Role of Retinal Glial Cells in Stem Cell-Based Therapy for Neuroretinal Diseases Not only that, a new strategy for stem cell-based combined transplantation has been developed to treat various no-ocular diseases or injury such as severe aplastic anemia [348], acute blood loss [349], Graft Versus Host Disease (GVHD) [350], myocardial infarction [351], diabetes [352], and cavernous nerve injury-related erectile dysfunction [33]. And these studies of combined transplantation confirmed a more positive and encouraging result than that of single transplantation. Previous studies have demonstrated that MSCs were able to promote the survival and functionalization of retinal cells after single transplantation through secretion of neuroprotective factors and deactivation of inflammatory pathway [354-356]. As expected, combined transplantation of human RPCs and MSCs into subretinal space of RCS rats presented better therapeutic effect in functional maintenance and structural integration than that of single transplantation [357]. Likewise, in vivo intravitreal combined transplantation of human iPSCs and mouse RGCs facilitated RGC neurites into longer extension than single transplantation of mouse RGCs [358]. Therefore, for treating neuroretinal diseases effectively, a new co transplantation tactics in combination with stem cell-derived photoreceptors or RPE cells appears to be a promising way to give full play to the effects of transplanted cells. Microglia is selected as targeted cells for combined transplantation with stem cell-derived photoreceptors or RPE cells. Microglia are a group of highly specialized immune cells and widely exist in CNS, including retina, which play a pivotal role in immune surveillance and maintaining homeostasis of retinal microenvironment [359,360]. Microglia has been confirmed to have neuroprotective effects against neuronal death in ischemic brain injury [361,362]. And microglia is also capable of phagocytizing injured photoreceptors and preventing the death of photoreceptors in acute retinal detachment [363]. Recent studies further demonstrated that subretinal microglia in photoreceptor degeneration played an important role in protection of RPE structural integrity [364]. Therefore, microglia should be able to improve subretinal microenvironment prior to transplantation suitable for the survival of transplanted photoreceptors or RPE cells. In addition, microglia can be generated from induction of human ESCs and iPSC [365,366], providing sufficient cell sources for combined transplantation studies. Taken together, combined transplantation of stem cell-derived microglia and photoreceptors or RPE cells will become the next another target to treat neuroretinal diseases. Moreover, as mentioned above, Müller Glia (MG) are also promising targeted cells to be directly reactivated and differentiate into new retinal neurons in vivo with the development of induced techniques. Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 9 of 22 •

Donor cell sources

Human MG-derived photoreceptors

Human RPCs and MSCs

Host animal models

Transgenic P23H rats [367]

RCS rats with Mertk mutation [368]

Target diseases

Cell dose

Retinitis Pigmentosa (RP)

Total 2 μl containing 1 μl cell suspension and 4 × 104 photoreceptor differentiated cells

RP

Rho−/− mice [373]

RP

Gucy2e-/-mice [371]

Leber congenital amaurosis (LCA)

Subretinal injection

Subretinal injection

Gnat1−/− mice [235]

2× 10 cells/1 μl sorted by FACS 5

CSNB

Rho−/− mice [373]

Reference

4 weeks after transplantation

Transplanted cells migrated and integrated into the ONL Significant rod function improvement measured by scotopic flash ERG

[80]

3 weeks after transplantation

Combined transplantation has a higher ratio of photoreceptor differentiation and low immunoreaction than single transplantation

[357]

3-6 months after transplantation

Preserve visual function measured by OKR

[376]

Subretinal injection via superior sclera

2-3 weeks after transplantation

No robust integration and maturation of photoreceptors Increased cell death and gliosis

[96]

8-12 weeks Subretinal injection by trans-scleral route

2× 10 cells/1 μl sorted by FACS

mice Prph2 [374]

Effects of transplantation

Congenital stationary night blindness(CSNB)

5

rd2/rd2

Time point for evaluation

6-12weeks

Rho−/− mice [379]

Mouse iPSC-derived photoreceptor precursors using 2D culture

3 weeks

8-12weeks Gnat-/- mice [235]

Mouse ESC-derived rod photoreceptors using 3D culture

3 weeks

Delivery methods

5× 104-1× 105 cells/2 μL

Human RPCs

Mouse ESC-derived rod photoreceptors using 2D culture

Total 4 × 105 cells/5 μl separately and together

Time window (postnatal)

3 weeks after transplantation

3-4 weeks RP

RP

4-6 week

Subretinal injection

3 weeks after transplantation

1.1.6 × 105 integrated cells 2. Increased b-wave amplitude measured by ERG

[224]

[239]

[129]

Mouse iPSC-derived retinal sheets using 3D culture

Pde6brd1 mouse [375]

RP

0.5 mm × 2 mm retinal sheets

6-9 weeks

Subretinal injection

2-4 weeks after transplantation

Photoreceptor responses recorded by ex vivo micro-ERG and ganglion cell responses recorded MEA

Mouse ESC/ iPSC derived retinal sheet using 3D culture

Pde6brd1 mouse [375]

RP

0.5 mm x 1.5-2 mm retinal sheets

6-8 weeks

Subretinal injection

2 weeks to 6 months after transplantation

Form synaptic connections

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

[372]

Form shorter segments

8 weeks

2.5× 105 cells/lμl

2.5 × 104 integrated cells form long outer segments Vision restoration measured by water maze and optometry system

Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 10 of 22 •

Mouse ESC-derived cone photoreceptors using 3D culture

Human ESC-derived photoreceptor precursors using 2D culture

Human ESC- and iPSC-derived photoreceptor progenitors

Nrl−/− mice [376]

“Cod” hybrid phenotype

Prph2rd2/rd2 [374]

RP

RPE65R91W/R91W [377]

Cone degeneration

Crx -/- mice [379]

Pde6brd1 mouse [375]

LCA

RP

Significant material transfer 2× 105 cells/1 μL sorted by FACS

Immunodeficient rho S334ter-3 rat [380]

5× 104-8× 104 cells/1 μL

2 × 105 cells/2μL

0-3 days (intravitreal) and 4-6 weeks (subretinal)

10-12 weeks

Intravitreal and subretinal injections

Subretinal injection by trans-scleral route

4,5,6,9,11 years

RP

Retinal sheets

26-38 days

Immunodeficient NOG-rd1-2J and NOG-rd10 [382]

Human iPSC-derived retinal sheets using 3D culture

Subretinal injection by trans-scleral route

Subretinal injection

8 weeks

Immunodeficient SD-Foxn1 Tg(S334ter)3LavRrrc nude rats [383]

2-5 months

RP

2-3 weeks after transplantation

Significant cones integration

[238]

Significant material transfer

RP-like monkey model induced by cobalt chloride or laser photocoagulation [313]

Human ESC-derived retinal sheets using 3D culture

2-3 months

[107]

3 weeks after transplantation

Survival and maturation in vivo of few transplanted cells Partial vision recovery measured by optomotor response (OMR) and light avoidance behavior

[109]

35, 88, 123, and 148 days after transplantation

Lack positive functional results Differentiation into various retinal cells

[313]

2 weeks after transplantation

Produce functional photoreceptor Visual improvements measured by OKR and SC electrophysiologic recording

[381]

4 weeks after transplantation

Long-term survival Functional integration Light responses measured by MEA

[382]

3-5 months after transplantation

Formation of outer segments and integration into host bipolar cells Observed RGC light responses by MEA

Subretinal injection by trans-vitreal route

Retinal sheets

RP-like monkey model induced by cobalt chloride or laser photocoagulation [313]

2 weeks after transplantation

1.3 × 103Nrl+ integrated human cells 2. A small, significant ERG response

9 months

6-7 months after transplantation

[252]

Mild vision recovery measured by Visually-Guided Saccades (VGS)

Table 1: Representative stem cell transplantation studies in animal models of photoreceptor-defective diseases.

Conclusion

Competing Interest

In terms of depth and breadth, translational medicine studies on stem cell transplantation for treatment of neuroretinal diseases have made considerable progress. Although these encouraging advances have brought great hope to the treatment of neuroretinal diseases, there are still big challenges ahead that need to be further explored and overcome. The new techniques of stem cell-based transplantation are also worthy of unwearied pursuit.

The author declares that has no competing interests.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

Acknowledgment This work was partly supported by Zhejiang Provincial Natural Science Foundation of China (LQ17H120005); National Key Research and Development Program of China (2017YFA0105300); Ministry of Education 111 project (D16011). Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 11 of 22 •

References 1.

Rodieck RW (1998) The First Steps in Seeing. Arch Ophthalmol 117: 550.

2.

Marshall J (1989) The retina: an approachable part of the brain. Br J Ophthalmol 73: 479-480.

3.

[No authors listed] (1999) The Human Eye: Structure and Function. Nat Med 5: 1229.

4.

Willoughby CE, Ponzin D, Ferrari S, Lobo A, Landau K, et al. (2010) Anatomy and physiology of the human eye: effects of mucopolysaccharidoses disease on structure and function - a review. Clin Exp Ophthalmol 38: 2-11.

5.

Johnson GJ, Minassian DC, Weale RA, West SK (2003) The Epidemiology of Eye Disease. Epidemiology of eye disease 58: 501.

6.

Klein R, Klein BE (2006) The epidemiology of eye disease: from glycemia to genetics: the Friedenwald Lecture. Invest Ophthalmol Vis Sci 47: 1746-1753.

7.

Cepko CL, Austin CP, Yang X, Alexiades M, Ezzeddine D (1996) Cell fate determination in the vertebrate retina. Proc Natl Acad Sci U S A 93: 589-595.

8.

Masland RH (2001) The fundamental plan of the retina. Nat Neurosci 4: 877-886.

9.

Livesey FJ, Cepko CL (2001) Vertebrate neural cell-fate determination: Lessons from the retina. Nat Rev Neurosci 2: 109-118.

10. Landau K, Kurz-Levin M (2011) Retinal disorders. Handb Clin Neurol 102: 97. 11. Marc RE (2010) Injury and Repair: Retinal Remodeling. Encyclopedia of the Eye Pg no: 414-420. 12. Ambati J, Fowler BJ (2012) Mechanisms of Age-Related Macular Degeneration. Neuron 75: 26-39. 13. de Jong PTVM (2006) Mechanisms of disease: Age-related macular degeneration. N Engl J Med, 355: 1474-1485. 14. Sparrow JR, Yamamoto K, Ablonczy Z, Gutierrez DB, Grey AC, et al. (2012) Retinal Degenerative Diseases. Advances in Experimental Medicine & Biology pg no: 854. 15. Hartong DT, Berson EL, Dryja TP (2006) Retinitis pigmentosa. Lancet 368: 1795-1809. 16. Geller AM, Sieving PA (1993) Assessment of foveal cone photoreceptors in Stargardt’s macular dystrophy using a small dot detection task. Vision Res 33: 1509-1524. 17. Berson EL (1993) Retinitis pigmentosa. The Friedenwald Lecture. Invest Ophthalmol Vis Sci 34: 1659-1676. 18. Bunker CH, Berson EL, Bromley WC, Hayes RP, Roderick TH (1984) Prevalence of retinitis pigmentosa in Maine. Am J Ophthalmol 97: 357365. 19. Waugh N, Loveman E, Colquitt J, Royle P, Yeong JL, et al. (2018) Treatments for dry age-related macular degeneration and Stargardt disease: a systematic review. Health Technol Assess 22: 1-168. 20. Facts about age-related macular degeneration. 21. Coleman HR, Chan CC, Ferris FL, Chew EY (2008) Age-related macular degeneration. Lancet 372: 1835-1845. 22. Schuman SG, Koreishi AF, Farsiu S, Jung SH, Izatt JA, et al. (2009) Photoreceptor layer thinning over drusen in eyes with age-related macular degeneration imaged in vivo with spectral-domain optical coherence tomography. Ophthalmology 116: 488-496. J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

23. Zeiss CJ (2010) Animals as models of age-related macular degeneration: an imperfect measure of the truth. Vet Pathol 47: 396-413. 24. Zarbin MA (2004) Current concepts in the pathogenesis of age-related macular degeneration. Arch Ophthalmol 122: 598-614. 25. Gehrs KM, Jackson JR, Brown EN, Allikmets R, Hageman GS (2010) Complement, age-related macular degeneration and a vision of the future. Arch Ophthalmol 128: 349-358. 26. Ding X, Patel M, Chan CC (2009) Molecular pathology of age-related macular degeneration. Prog Retin Eye Res 28: 1-18. 27. Owen CG, Jarrar Z, Wormald R, Cook DG, Fletcher AE, et al. (2012) The estimated prevalence and incidence of late stage age related macular degeneration in the UK. Br J Ophthalmol 96: 752-756. 28. Wong WL, Su X, Li X, Cheung CM, Klein R, et al. (2014) Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: a systematic review and meta-analysis. Lancet Glob Health 2: 106-116. 29. Day S, Acquah K, Lee PP, Mruthyunjaya P, Sloan FA (2011) Medicare costs for neovascular age-related macular degeneration, 1994-2007. Am J Ophthalmol 152: 1014-1020. 30. Michaelides M, Hunt D, Moore A (2003) The genetics of inherited macular dystrophies. J Med Genet 40: 641-650. 31. Fujinami K, Lois N, Davidson AE, Mackay DS, Hogg CR, et al. (2013) A longitudinal study of stargardt disease: clinical and electrophysiologic assessment, progression, and genotype correlations. Am J Ophthalmol 155: 1075-1088. 32. Strauss RW, Ho A, Muñoz B, Cideciyan AV, Sahel JA, et al. (2016) The Natural History of the Progression of Atrophy Secondary to Stargardt Disease (ProgStar) Studies: Design and Baseline Characteristics: ProgStar Report No. 1. Ophthalmology 123: 817-828. 33. Lambertus S, van Huet RA, Bax NM, Hoefsloot LH, Cremers FP, et al. (2015) Early-onset stargardt disease: phenotypic and genotypic characteristics. Ophthalmology 122: 335-344. 34. Chiba C, Hyde DR, Vihtelic TS, Fimbel SM, Kassen SC, et al. (2005) [Strategies for Retinal Regeneration in Vertebrates: from Fish to Human]. Zool Sci 22: 1373-1376. 35. Grigoryan EN (2018) Endogenous Cell Sources for Eye Retina Regeneration in Vertebrate Animals and Humans. Russ J Dev Biol 49: 314-326. 36. Stern JH, Tian Y, Funderburgh J, Pellegrini G, Zhang K, et al. (2018) Regenerating Eye Tissues to Preserve and Restore Vision. Cell Stem Cell 22: 834-849. 37. Trounson A, McDonald C (2015) Stem Cell Therapies in Clinical Trials: Progress and Challenges. Cell Stem Cell 17: 11-22. 38. Zamiri P, Sugita S, Streilein JW (2007) Immunosuppressive properties of the pigmented epithelial cells and the subretinal space. Chem Immunol Allergy 92: 86-93. 39. Jiang LQ, Jorquera M, Streilein JW (1993) Subretinal space and vitreous cavity as immunologically privileged sites for retinal allografts. Invest Ophthalmol Vis Sci 34: 3347-3354. 40. Caspi RR (2010) A look at autoimmunity and inflammation in the eye. J Clin Invest 120: 3073-3083. 41. Willett K, Bennett J (2013) Immunology of AAV-Mediated Gene Transfer in the Eye. Front Immunol 4: 261. 42. Royo PE, Quay WB (1959) Retinal transplantation from fetal to maternal mammalian eye. Growth 23: 313-336. 43. Cepko C (2014) Intrinsically different retinal progenitor cells produce specific types of progeny. Nat Rev Neurosci 15: 615-627. Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 12 of 22 •

44. Tropepe V, Coles BL, Chiasson BJ, Horsford DJ, Elia AJ, et al. (2000) Retinal stem cells in the adult mammalian eye. Science 287: 2032-2036.

63. Reichenbach A, Bringmann A (2013) New functions of Müller cells. Glia 61: 651-678.

45. Reh TA, Levine EM (1999) Multipotential stem cells and progenitors in the vertebrate retina. J Neurobiol 36: 206-220.

64. Bringmann A, Pannicke T, Biedermann B, Francke M, Iandiev I, et al. (2009) Role of retinal glial cells in neurotransmitter uptake and metabolism. Neurochem Int 54: 143-160.

46. Klassen HJ, Ng TF, Kurimoto Y, Kirov I, Shatos M, et al. (2004) Multipotent retinal progenitors express developmental markers, differentiate into retinal neurons, and preserve light-mediated behavior. Invest Ophthalmol Vis Sci 45: 4167-4173.

65. Newman E, Reichenbach A (1996) The Müller cell: a functional element of the retina. Trends Neurosci 19: 307-312.

47. Luo J, Baranov P, Patel S, Ouyang H, Quach J, et al. (2014) Human retinal progenitor cell transplantation preserves vision. J Biol Chem 289: 6362-6371.

66. Ogai K, Hisano S, Sugitani K, Koriyama Y, Kato S (2016) Cell Fate of Müller Cells During Photoreceptor Regeneration in an N-Methyl-N-nitrosourea-Induced Retinal Degeneration Model of Zebrafish. Adv Exp Med Biol 854: 685-692.

48. Chacko DM, Rogers JA, Turner JE, Ahmad I (2000) Survival and differentiation of cultured retinal progenitors transplanted in the subretinal space of the rat. Biochem Biophys Res Commun 268: 842-846.

67. Fischer AJ, Reh TA (2001) Müller glia are a potential source of neural regeneration in the postnatal chicken retina. Nat Neurosci 4: 247-252.

49. Turner DL, Cepko CL (1987) A common progenitor for neurons and glia persists in rat retina late in development. Nature 328: 131-136.

68. Fausett BV, Goldman D (2006) A role for alpha1 tubulin-expressing Müller glia in regeneration of the injured zebrafish retina. J Neurosci 26: 6303-6313.

50. MacLaren RE, Pearson RA, MacNeil A, Douglas RH, Salt TE, et al. (2006) Retinal repair by transplantation of photoreceptor precursors. Nature 444: 203-207.

69. Vecino E, Rodriguez FD, Ruzafa N, Pereiro X, Sharma SC (2016) Glia-neuron interactions in the mammalian retina. Prog Retin Eye Res 51: 1-40.

51. Qu Z, Guan Y, Cui L, Song J, Gu J, et al. (2015) Transplantation of rat embryonic stem cell-derived retinal progenitor cells preserves the retinal structure and function in rat retinal degeneration. Stem Cell Res Ther 6: 219.

70. Ooto S, Akagi T, Kageyama R, Akita J, Mandai M, et al. (2004) Potential for neural regeneration after neurotoxic injury in the adult mammalian retina. Proc Natl Acad Sci U S A 101: 13654-13659.

52. Klassen H, Kiilgaard JF, Warfvinge K, Samuel MS, Prather RS, et al. (2012) Photoreceptor Differentiation following Transplantation of Allogeneic Retinal Progenitor Cells to the Dystrophic Rhodopsin Pro347Leu Transgenic Pig. Stem Cells Int 2012: 939801. 53. Mayer EJ, Carter DA, Ren Y, Hughes EH, Rice CM, et al. (2005) Neural progenitor cells from postmortem adult human retina. Br J Ophthalmol 89: 102-106. 54. Mayer EJ, Hughes EH, Carter DA, Dick AD (2003) Nestin positive cells in adult human retina and in epiretinal membranes. Br J Ophthalmol 87: 1154-1158. 55. Qiu G, Seiler MJ, Thomas BB, Wu K, Radosevich M, et al. (2007) Revisiting nestin expression in retinal progenitor cells in vitro and after transplantation in vivo. Exp Eye Res 84: 1047-1059. 56. Cho SJ, Lee YS, Lee JI, Bang JI, Yang J, et al. (2010) Confirmation of germ-line transmission in the red fluorescence protein (RFP) transgenic cloned male cat. Cell Reprogram 12: 739-747. 57. Klassen H, Kiilgaard JF, Zahir T, Ziaeian B, Kirov I, et al. (2007) Progenitor cells from the porcine neural retina express photoreceptor markers after transplantation to the subretinal space of allorecipients. Stem cells 25: 1222-1230. 58. Aftab U, Jiang C, Tucker B, Kim JY, Klassen H, et al. (2009) Growth kinetics and transplantation of human retinal progenitor cells. Exp Eye Res 89: 301-310. 59. Li SY, Yin ZQ, Chen SJ, Chen LF, Liu Y (2009) Rescue from light-induced retinal degeneration by human fetal retinal transplantation in minipigs. Curr Eye Res 34: 523-535. 60. Hamon A, Roger JE, Yang XJ, Perron M: Müller glial cell-dependent regeneration of the neural retina: An overview across vertebrate model systems. Dev Dyn 245: 727-738.

71. Karl MO, Hayes S, Nelson BR, Tan K, Buckingham B, et al. (2008) Stimulation of neural regeneration in the mouse retina. Proc Natl Acad Sci U S A 105: 19508-19513. 72. Ueki Y, Reh TA, (2013) EGF stimulates Müller glial proliferation via a BMP-dependent mechanism. Glia 61: 778-789. 73. Wan J, Zheng H, Chen ZL, Xiao HL, Shen ZJ, et al. (2008) Preferential regeneration of photoreceptor from Müller glia after retinal degeneration in adult rat. Vision Res 48: 223-234. 74. Wohl SG, Schmeer CW, Kretz A, Witte OW, Isenmann S (2009) Optic nerve lesion increases cell proliferation and nestin expression in the adult mouse eye in vivo. Exp Neurol 219: 175-186. 75. Tackenberg MA, Tucker BA, Swift JS, Jiang C, Redenti S, et al. (2009) Müller cell activation, proliferation and migration following laser injury. Mol Vis 15: 1886-1896. 76. Otteson DC, Phillips MJ (2010) A conditional immortalized mouse muller glial cell line expressing glial and retinal stem cell genes. Invest Ophthalmol Vis Sci 51: 5991-6000. 77. Giannelli SG, Demontis GC, Pertile G, Rama P, Broccoli V (2011) Adult human Müller glia cells are a highly efficient source of rod photoreceptors. Stem cells 29: 344-356. 78. Wan J, Zheng H, Xiao HL, She ZJ, Zhou GM (2007) Sonic hedgehog promotes stem-cell potential of Müller glia in the mammalian retina. Biochem Biophys Res Commun 363: 347-354. 79. Singhal S, Bhatia B, Jayaram H, Becker S, Jones MF, et al. (2012) Human Müller glia with stem cell characteristics differentiate into retinal ganglion cell (RGC) precursors in vitro and partially restore RGC function in vivo following transplantation. Stem Cells Transl Med 1: 188199.

61. Goldman D (2014) Müller glial cell reprogramming and retina regeneration. Nat Rev Neurosci 15: 431-442.

80. Jayaram H, Jones MF, Eastlake K, Cottrill PB, Becker S, et al. (2014) Transplantation of photoreceptors derived from human Muller glia restore rod function in the P23H rat. Stem Cells Transl Med 3: 323-333.

62. Jorstad NL, Wilken MS, Grimes WN, Wohl SG, VandenBosch LS, et al. (2017) Stimulation of functional neuronal regeneration from Müller glia in adult mice. Nature 548: 103-107.

81. Salero E, Blenkinsop TA, Corneo B, Harris A, Rabin D, et al. (2012) Adult human RPE can be activated into a multipotent stem cell that produces mesenchymal derivatives. Cell Stem Cell 10: 88-95.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 13 of 22 •

82. Saini JS, Temple S, Stern JH (2016) Human Retinal Pigment Epithelium Stem Cell (RPESC). Adv Exp Med Biol 854: 557-562. 83. Miyake A, Araki M (2014) Retinal stem/progenitor cells in the ciliary marginal zone complete retinal regeneration: a study of retinal regeneration in a novel animal model. Dev Neurobiol 74: 739-756. 84. Bhatia B, Jayaram H, Singhal S, Jones MF, Limb GA (2011) Differences between the neurogenic and proliferative abilities of Müller glia with stem cell characteristics and the ciliary epithelium from the adult human eye. Exp Eye Res 93: 852-861. 85. Jasty S, Srinivasan P, Pasricha G, Chatterjee N, Subramanian K (2012) Gene expression profiles and retinal potential of stem/progenitor cells derived from human iris and ciliary pigment epithelium. Stem Cell Rev 8: 1163-1177. 86. Frøen RC, Johnsen EO, Petrovski G, Berényi E, Facskó A, et al. (2011) Pigment epithelial cells isolated from human peripheral iridectomies have limited properties of retinal stem cells. Acta Ophthalmol 89: 635644. 87. Cai H, Shin MC, Tezel TH, Kaplan HJ, Del Priore LV (2006) Use of iris pigment epithelium to replace retinal pigment epithelium in age-related macular degeneration: a gene expression analysis. Arch Ophthalmol 124: 1276-1285.

100. Kobayashi W, Onishi A, Tu HY, Takihara Y, Matsumura M, et al. (2018) Culture Systems of Dissociated Mouse and Human Pluripotent Stem Cell-Derived Retinal Ganglion Cells Purified by Two-Step Immunopanning. Invest Ophthalmol Vis Sci 59: 776-787. 101. Bertacchi M, Lupo G, Pandolfini L, Casarosa S, D’Onofrio M, et al. (2015) Activin/Nodal Signaling Supports Retinal Progenitor Specification in a Narrow Time Window during Pluripotent Stem Cell Neuralization. Stem Cell Rep 5:532-545. 102. Mellough CB, Collin J, Khazim M, White K, Sernagor E, et al. (2015) IGF-1 Signaling Plays an Important Role in the Formation of Three-Dimensional Laminated Neural Retina and Other Ocular Structures From Human Embryonic Stem Cells. Stem Cells33:2416-2430. 103. Banin E, Obolensky A, Idelson M, Hemo I, Reinhardtz E, et al. (2006) Retinal incorporation and differentiation of neural precursors derived from human embryonic stem cells. Stem Cells 24:246-257. 104. Boucherie C, Mukherjee S, Henckaerts E, Thrasher AJ, Sowden JC, et al. (2013) Brief report: self-organizing neuroepithelium from human pluripotent stem cells facilitates derivation of photoreceptors. 31:408-414. 105. Hirami Y, Osakada F, Takahashi K, Okita K, Yamanaka S, et al. (2009) Generation of retinal cells from mouse and human induced pluripotent stem cells. Neurosci Lett 458:126-131.

88. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, et al. (1998) Embryonic stem cell lines zderived from human blastocysts. Science 282: 1145-1147.

106. Ikeda H, Osakada F, Watanabe K, Mizuseki K, Haraguchi T, et al. (2005) Generation of Rx+/Pax6+ neural retinal precursors from embryonic stem cells.Proc Natl Acad Sci U S A 102: 11331-11336.

89. Takahashi K, Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126: 663-676.

107. Lamba DA, Gust J, Reh TA (2009) Transplantation of human embryonic stem cell-derived photoreceptors restores some visual function in Crx-deficient mice.Cell Stem Cell 4: 73-79.

90. Uccelli A, Moretta L, Pistoia V (2008) Mesenchymal stem cells in health and disease. Nat Rev Immunol 8: 726-736.

108. Osakada F1, Ikeda H, Mandai M, Wataya T, Watanabe K, et al. (2008) Toward the generation of rod and cone photoreceptors from mouse, monkey and human embryonic stem cells. Nat Biotechnol 26: 215-224.

91. Rezanejad H, Soheili ZS, Haddad F, Matin MM, Samiei S, et al. (2014) In vitro differentiation of adipose-tissue-derived mesenchymal stem cells into neural retinal cells through expression of human PAX6 (5a) gene. Cell Tissue Res 356: 65-75. 92. Bailo M, Soncini M, Vertua E, Signoroni PB, Sanzone S, et al. (2004) Engraftment potential of human amnion and chorion cells derived from term placenta. Transplantation 78: 1439-1448. 93. Tzameret A, Sher I, Belkin M, Treves AJ, Meir A, et al. (2015) Epiretinal transplantation of human bone marrow mesenchymal stem cells rescues retinal and vision function in a rat model of retinal degeneration. Stem Cell Res 15: 387-394. 94. Hartmann K, Raabe O, Wenisch S, Arnhold S (2013) Amniotic fluid derived stem cells give rise to neuron-like cells without a further differentiation potential into retina-like cells. Am J Stem Cells 2: 108-118. 95. Martin GR (1981) Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad Sci USA 78: 7634-7638. 96. West EL, Gonzalez-Cordero A, Hippert C, Osakada F, Martinez-Barbera JP, et al. (2012) Defining the Integration Capacity of Embryonic Stem Cell-Derived Photoreceptor Precursors. Stem Cells 30: 1424-1435. 97. Tucker BA, Mullins RF, Streb LM, Anfinson K, Eyestone ME, et al. (2013) Patient-specific iPSC-derived photoreceptor precursor cells as a means to investigate retinitis pigmentosa. Elife 2: 00824.

109. Barnea-Cramer AO, Wang W, Lu SJ, Singh MS, Luo C, et al. (2016) Function of human pluripotent stem cell-derived photoreceptor progenitors in blind mice. Sci Rep 6: 29784. 110. Zhao X, Liu J, Ahmad I (2002) Differentiation of embryonic stem cells into retinal neurons. Biochem Biophys Res Commun 297: 177-184. 111. Banin E, Obolensky A, Idelson M, Hemo I, Reinhardtz E, et al. (2006) Retinal incorporation and differentiation of neural precursors derived from human embryonic stem cells. Stem Cells 24: 246-257. 112. Lamba DA, Karl MO, Ware CB, Reh TA (2006) Efficient generation of retinal progenitor cells from human embryonic stem cells. Proc Natl Acad Sci U S A 103: 12769-12774. 113. Osakada F, Ikeda H, Mandai M, Wataya T, Watanabe K, et al. (2008) Toward the generation of rod and cone photoreceptors from mouse, monkey and human embryonic stem cells. Nat Biotechno l26: 215-224. 114. Ringdén O, Le Blanc K, Hovatta O (2003) Transplantation of embryonic stem cells: possibilities and challenges. Transplantation 76: 1011-1012. 115. Yamanaka S (2007) Strategies and new developments in the generation of patient-specific pluripotent stem cells. Cell Stem Cell 1: 39-49. 116. Osakada F, Ikeda H, Sasai Y, Takahashi M (2009) Stepwise differentiation of pluripotent stem cells into retinal cells. Nat Protoc 4: 811-824.

98. Sowden JC (2014) ESC-derived retinal pigmented epithelial cell transplants in patients: so far, so good. Cell Stem Cell 15: 537-538.

117. Phillips MJ, Wallace KA, Dickerson SJ, Miller MJ, Verhoeven AD, et al. (2012) Blood-derived human iPS cells generate optic vesicle-like structures with the capacity to form retinal laminae and develop synapses. Invest Ophthalmol Vis Sci 53: 2007-2019.

99. Mandai M, Watanabe A, Kurimoto Y, Hirami Y, Morinaga C, et al. (2017) Autologous Induced Stem-Cell-Derived Retinal Cells for Macular Degeneration. N Engl J Med 376: 1038-1046.

118. Meyers JR, Hu L, Moses A, Kaboli K, Papandrea A, et al. (2012) β-catenin/Wnt signaling controls progenitor fate in the developing and regenerating zebrafish retina. Neural Dev 7: 30.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 14 of 22 •

119. Amirpour N, Karamali F, Rabiee F, Rezaei L, Esfandiari E, et al. (2012) Differentiation of human embryonic stem cell-derived retinal progenitors into retinal cells by Sonic hedgehog and/or retinal pigmented epithelium and transplantation into the subretinal space of sodium iodate-injected rabbits. Stem CellsDev 21: 42-53.

135. Santos-Ferreira T, Völkner M, Borsch O, Haas J, Cimalla P, et al. (2016) Stem Cell-Derived Photoreceptor Transplants Differentially Integrate Into Mouse Models of Cone-Rod Dystrophy. Invest Ophthalmol Vis Sci 57:3509-3520.

120. Mellough CB, Sernagor E, Moreno-Gimeno I, Steel DH, Lako M, et al. (2012) Efficient stage-specific differentiation of human pluripotent stem cells toward retinal photoreceptor cells. Stem Cells 30: 673-686.

136. Ohlemacher SK, Iglesias CL, Sridhar A, Gamm DM, Meyer JS (2015) Generation of highly enriched populations of optic vesicle-like retinal cells from human pluripotent stem cells. Curr Protoc Stem Cell Biol 32: 1-20.

121. Tucker BA, Mullins RF, Streb LM, Anfinson K, Eyestone ME, et al. (2013) Patient-specific iPSC-derived photoreceptor precursor cells as a means to investigate retinitis pigmentosa. Elife 2: 00824.

137. Mandai M, Watanabe A, Kurimoto Y, Hirami Y, Morinaga C (2017) Autologous Induced Stem-Cell-Derived Retinal Cells for Macular Degeneration. N Engl J Med 376:1038-1046.

122. Yanai A, Laver CR, Joe AW, Viringipurampeer IA, Wang X, et al. (2013) Differentiation of human embryonic stem cells using size-controlled embryoid bodies and negative cell selection in the production of photoreceptor precursor cells. Tissue Eng Part C Methods 19: 755-764.

138. Lowe A, Harris R, Bhansali P, Cvekl A, Liu W ((2016) Intercellular Adhesion-Dependent Cell Survival and ROCK-Regulated Actomyosin-Driven Forces Mediate Self-Formation of a Retinal Organoid. Stem Cell Rep6:743-756.

123. Zhou S, Flamier A, Abdouh M, Tétreault N, Barabino, et al. (2019) Differentiation of human embryonic stem cells into cone photoreceptors through simultaneous inhibition of BMP, TGFβ and Wnt signaling. Development 142: 3294-3306.

139. Kruczek K, Gonzalez-Cordero A, Goh D, Naeem A, Jonikas M, et al. (2017) Differentiation and transplantation of embryonic stem cell-derived cone photoreceptors into a mouse model of end-stage retinal degeneration. Stem Cell Rep 8: 1659-1674.

124. Eiraku M, Takata N, Ishibashi H, Kawada M, Sakakura E, et al. (2011) Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature 472: 51-56.

140. Hiler D, Chen X, Hazen J, Kupriyanov S, Carroll PA, et al. (2015) Quantification of retinogenesis in 3D cultures reveals epigenetic memory and higher efficiency in iPSCS derived from rod photoreceptors. Cell Stem Cell 17: 101-115.

125. Nakano T, Ando S, Takata N, Kawada M, Muguruma K, et al. (2012) Self-formation of optic cups and storable stratified neural retina from human ESCs.Cell Stem Cell 10: 771-785. 126. Phillips MJ, Perez ET, Martin JM, Reshel ST, Wallace KA, et al. (2014) Modeling human retinal development with patient-specific induced pluripotent stem cells reveals multiple roles for visual system homeobox 2. Stem Cells 32: 1480-1492. 127. Reichman S, Slembrouck A, Gagliardi G, Chaffiol A, Terray A, et al. (2017) Generation of Storable Retinal Organoids and Retinal Pigmented Epithelium from Adherent Human iPS Cells in Xeno-Free and Feeder-Free Conditions. Stem Cells 35: 1176-1188. 128. Wahlin K, Maruotti JA, Sripathi SR, Ball J, Angueyra JM, et al. (2017) Photoreceptor Outer Segment-like Structures in Long-Term 3D Retinas from Human Pluripotent Stem Cells. Sci Rep 7: 766. 129. Assawachananont J, Mandai M, Okamoto S, Yamada C, Eiraku M, et al. (2014) Transplantation of embryonic and induced pluripotent stem cell-derived 3D retinal sheets into retinal degenerative mice. Stem Cell Rep 2: 662-674. 130. Ben M’Barek K, Habeler W, Plancheron A, Jarraya M, Regent F, et al. (2017) Human ESC-derived retinal epithelial cell sheets potentiate rescue of photoreceptor cell loss in rats with retinal degeneration. Sci Transl Med 9. 131. Chen HY, Kaya KD, Dong L, Swaroop A (2016) Three-dimensional retinal organoids from mouse pluripotent stem cells mimic development with enhanced stratification and rod photoreceptor differentiation. Mol Vis 22: 1077-1094.

141. Hiler DJ, Barabas ME, Griffiths LM, Dyer MA (2016) Reprogramming of mouse retinal neurons and standardized quantification of their differentiation in 3D retinal cultures. Nat Protoc 11: 1955-1976. 142. Gonzalez-Cordero A, Kruczek K, Naeem A, Fernando M, Kloc M, et al. (2017) Recapitulation of human retinal development from human pluripotent stem cells generates transplantable populations of cone photoreceptors. Stem Cell Rep 9: 820-837. 143. Llonch S, Carido M, Ader M (2018) Organoid technology for retinal repair. Dev Biol 433: 132-143. 144. Kuwahara A, Ozone C, Nakano T, Saito K, Eiraku M, et al. (2015) Generation of a ciliary margin-like stem cell niche from self-organizing human retinal tissue. Nat Commun 6: 6286. 145. Capowski EE, Samimi K, Mayerl SJ, Phillips MJ, Pinilla I, et al. (2019) Reproducibility and staging of 3D human retinal organoids across multiple pluripotent stem cell lines. Development 146. 146. Parfitt DA, Lane A, Ramsden CM, Carr A-JF, Munro PM, (2016) Identification and correction of mechanisms underlying inherited blindness in human iPSC-derived optic cups. Cell Stem Cell 18: 769-781. 147. Dutta D, Heo I, Clevers H (2017) Disease modeling in stem cell-derived 3d organoid systems. Trends Mol Med 23: 393-410. 148. Aparicio JG, Shayler DWH, Cobrinik D (2017) Retinal organoids: An emerging technology for retinal disease research and therapy. Cellular Therapies for Retinal Disease 117-138.

132. Decembrini S, Koch U, Radtke F, Moulin A, Arsenijevic Y (2014) Derivation of Traceable and Transplantable Photoreceptors from Mouse Embryonic Stem Cells. Stem Cell Rep 2: 853-865.

149. Deng WL, Gao ML, Lei XL, Lv JN, Zhao H, et al. (2018) Gene correction reverses ciliopathy and photoreceptor loss in iPSC-derived retinal organoids from retinitis pigmentosa patients. Stem Cell Rep 10: 12671281.

133. Zhong X, Gutierrez C, Xue T, Hampton C, Vergara MN, et al. (2014) : Generation of three-dimensional retinal tissue with functional photoreceptors from human iPSCs. Nat Commun 5:4047.

150. Yamane T, Hayashi S, Mizoguchi M, Yamazaki H, Kunisada T (1999) Derivation of melanocytes from embryonic stem cells in culture. Dev Dyn 216: 450-458.

134. Völkner M, Zschätzsch M, Rostovskaya M, Overall RW, Busskamp V, et al. (2016) Retinal Organoids from Pluripotent Stem Cells Efficiently Recapitulate Retinogenesis. Stem cell Rep 6:525-538.

151. Hirano M, Yamamoto A, Yoshimura N, Tokunaga T, Motohashi T, et al. (2003) Generation of structures formed by lens and retinal cells differentiating from embryonic stem cells. Dev Dyn 228: 664-671.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 15 of 22 •

152. Kawasaki H, Suemori H, Mizuseki K, Watanabe K, Urano F, et al. (2002) Generation of dopaminergic neurons and pigmented epithelia from primate ES cells by stromal cell-derived inducing activity. Proc Natl Acad Sci U S A 99: 1580-1585.

170. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, et al. (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131: 861-872.

153. Ooto S, Haruta M, Honda Y, Kawasaki H, Sasai Y, et al. (2003) Induction of the differentiation of lentoids from primate embryonic stem cells. Invest Ophthalmol Vis Sci 44: 2689-2693.

171. Buchholz DE, Pennington BO, Croze RH, Hinman CR, Coffey PJ, et al. (2013) Rapid and Efficient Directed Differentiation of Human Pluripotent Stem Cells Into Retinal Pigmented Epithelium. Stem Cells Transl Med 2: 384-393.

154. Vugler A, Carr A-J, Lawrence J, Chen LL, Burrell K, et al. (2008) Elucidating the phenomenon of HESC-derived RPE: Anatomy of cell genesis, expansion and retinal transplantation. Exp Neurol 214: 347-361.

172. Song MK, Lui GM (1990) Propagation of fetal human RPE cells: preservation of original culture morphology after serial passage. J Cell Physiol 143: 196-203.

155. Lund RD, Wang S, Klimanskaya I, Holmes T, Ramos-Kelsey R, et al. (2006) Human embryonic stem cell-derived cells rescue visual function in dystrophic RCS rats. Cloning Stem Cells 8: 189-199.

173. Rowland TJ, Blaschke AJ, Buchholz DE, Hikita ST, Johnson LV, et al (2013) Differentiation of human pluripotent stem cells to retinal pigmented epithelium in defined conditions using purified extracellular matrix proteins. J Tissue Eng Regen Med 7: 642-653.

156. Rowland TJ, Buchholz DE, Clegg DO (2012) Pluripotent human stem cells for the treatment of retinal disease. J Cell Physiol 227: 457-466. 157. Bharti K, Miller SS, Arnheiter H (2011) The new paradigm: Retinal pigment epithelium cells generated from embryonic or induced pluripotent stem cells. Pigment Cell Melanoma Res 24: 21-34. 158. Gong J, Sagiv O, Cai H, Tsang SH, Del Priore LV (2008) Effects of extracellular matrix and neighboring cells on induction of human embryonic stem cells into retinal or retinal pigment epithelial progenitors. Exp Eye Res 86: 957-965.

174. Hirami Y, Osakada F, Takahashi K, Okita K, Yamanaka S, et al. (2009) Generation of retinal cells from mouse and human induced pluripotent stem cells. Neurosci Lett 458: 126-131. 175. Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, et al. (2007) In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448: 318-324. 176. Yu J Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, et al. (2007) Induced pluripotent stem cell lines derived from human somatic cells. Science 318: 1917-1920.

159. Vugler A, Carr AJ, Lawrence J, Chen LL, Burrell K, et al. (2008) Elucidating the phenomenon of HESC-derived RPE: anatomy of cell genesis, expansion and retinal transplantation. Exp Neurol 214: 347-361.

177. Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka T, et al. (2008) Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol 26: 101-106.

160. Buchholz DE, Hikita ST, Rowland TJ, Friedrich AM, Hinman CR, et al. (2009) Derivation of functional retinal pigmented epithelium from induced pluripotent stem cells. Stem Cells 27: 2427-2434.

178. Banin E, Obolensky A, Idelson M, Hemo I, Reinhardtz E, et al. (2006) Retinal incorporation and differentiation of neural precursors derived from human embryonic stem cells. Stem Cells 24: 246-257.

161. Carr AJ, Vugler A, Lawrence J, Chen LL, Ahmado A, et al. (2009) Molecular characterization and functional analysis of phagocytosis by human embryonic stem cell-derived RPE cells using a novel human retinal assay. Mol Vis 15: 283-295.

179. Lu B, Morgans CW, Girman S, Luo J, Zhao J, et al. (2013) Neural Stem Cells Derived by Small Molecules Preserve Vision. Transl Vis Sci Technol 2: 1.

162. Lu B, Malcuit C, Wang S, Girman S, Francis P, et al. (2009) Long-term safety and function of RPE from human embryonic stem cells in preclinical models of macular degeneration. Stem Cells 27: 2126-2135. 163. Nistor G, Seiler MJ, Yan F, Ferguson D, Keirstead HS (2010) Three-dimensional early retinal progenitor 3D tissue constructs derived from human embryonic stem cells. J Neurosci Methods 190: 63-70. 164. Liao J-L, Yu J, Huang K, Hu J, Diemer T, et al. (2010) Molecular signature of primary retinal pigment epithelium and stem-cell-derived RPE cells. Hum Mol Genet 19: 4229-4238. 165. Osakada F, Jin Z-B, Hirami Y, Ikeda H, Danjyo T, (2009) In vitro differentiation of retinal cells from human pluripotent stem cells by small-molecule induction. J Cell Sci 122: 3169-3179. 166. Idelson M, Alper R, Obolensky A, Ben-Shushan E, Hemo I, et al. (2009) Directed Differentiation of Human Embryonic Stem Cells into Functional Retinal Pigment Epithelium Cells. Cell Stem Cell 5: 396-408. 167. Meyer JS, Shearer RL, Capowski EE, Wright LS, Wallace KA, et al. (2009) Modeling early retinal development with human embryonic and induced pluripotent stem cells. Proc Natl Acad Sci USA 106: 1669816703. 168. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, et al. (1998) Embryonic stem cell lines derived from human blastocysts. Science 282: 1145-1147. 169. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, et al. (2007) Induced pluripotent stem cell lines derived from human somatic cells. Science 318: 1917-1920.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

180. Tsai Y, Lu B, Bakondi B, Girman S, Sahabian A, et al. (2015) Human iPSC-Derived Neural Progenitors Preserve Vision in an AMD-Like Model. Stem Cells 33: 2537-2549. 181. Li L, Turner JE (1991) Optimal conditions for long-term photoreceptor cell rescue in RCS rats: the necessity for healthy RPE transplants. Exp Eye Res 52: 669-679. 182. Binder S, Stolba U, Krebs I, Kellner L, Jahn C, et al. (2002) Transplantation of autologous retinal pigment epithelium in eyes with foveal neovascularization resulting from age-related macular degeneration: a pilot study. Am J Ophthalmol 133: 215-225. 183. Heussen FM1, Fawzy NF, Joeres S, Lux A, Maaijwee K, et al. (2008) Autologous translocation of the choroid and RPE in age-related macular degeneration: 1-year follow-up in 30 patients and recommendations for patient selection. Eye (Lond) 22: 799-807. 184. Coffey PJ, Girman S, Wang SM, Hetherington L, Keegan DJ, et al. (2002) Long-term preservation of cortically dependent visual function in RCS rats by transplantation. Nat Neurosci 5: 53-56. 185. Carr AJ, Vugler AA, Hikita ST, Lawrence JM, Gias C, et al. (2009) Protective effects of human iPS-derived retinal pigment epithelium cell transplantation in the retinal dystrophic rat. PloS One 4: e8152. 186. Alvarez Palomo AB, McLenachan S, Chen FK, Da Cruz L, Dilley RJ, et al. (2015) Prospects for clinical use of reprogrammed cells for autologous treatment of macular degeneration. Fibrogenesis Tissue Repair 8: 9. 187. Blenkinsop TA, Saini JS, Maminishkis A, Bharti K, Wan Q, et al. (2015) Human Adult Retinal Pigment Epithelial Stem Cell-Derived RPE Monolayers Exhibit Key Physiological Characteristics of Native Tissue. Invest Ophthalmol Vis Sci 56: 7085-7099. Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 16 of 22 •

188. Blenkinsop TA, Salero E, Stern JH, Temple S (2013) The culture and maintenance of functional retinal pigment epithelial monolayers from adult human eye. Methods Mol Biol 945: 45-65. 189. Bharti K, Miller SS, Arnheiter H (2011) The new paradigm: retinal pigment epithelium cells generated from embryonic or induced pluripotent stem cells. Pigment Cell Melanoma Res 24: 21-34. 190. Singh R, Phillips MJ, Kuai D, Meyer J, Martin JM, et al. (2013) Functional analysis of serially expanded human iPS cell-derived RPE cultures. Invest Ophthalmol Vis Sci 54: 6767-6778. 191. van Meurs JC, Van Den Biesen PR (2003) Autologous retinal pigment epithelium and choroid translocation in patients with exudative age-related macular degeneration: short-term follow-up. Am J Ophthalmol 136: 688-695. 192. MacLaren RE, Uppal GS, Balaggan KS, Tufail A, Munro PM, et al. (2007) Autologous transplantation of the retinal pigment epithelium and choroid in the treatment of neovascular age-related macular degeneration. Ophthalmology 114: 561-570. 193. Treumer F, Bunse A, Klatt C, Roider J (2007) Autologous retinal pigment epithelium-choroid sheet transplantation in age related macular degeneration: morphological and functional results. Br J Ophthalmol 91:349-353. 194. Whiting P, Kerby J, Coffey P, da Cruz L, McKernan R (2015) Progressing a human embryonic stem-cell-based regenerative medicine therapy towards the clinic. Philos Trans R Soc Lond B Biol Sci 370: 20140375. 195. Machemer R, Steinhorst UH (1993) Retinal separation, retinotomy, and macular relocation: II. A surgical approach for age-related macular degeneration? Graefes Arch Clin Exp Ophthalmol 231: 635-641. 196. Del Priore LV, Kaplan HJ, Tezel TH, Hayashi N, Berger AS, et al. (2001) Retinal pigment epithelial cell transplantation after subfoveal membranectomy in age-related macular degeneration: Clinicopathologic correlation. Am J Ophthalmol 131: 472-480. 197. Buchholz DE, Hikita ST, Rowland TJ, Friedrich AM, Hinman CR, et al. (2009) Derivation of Functional Retinal Pigmented Epithelium from Induced Pluripotent Stem Cells. Stem Cells 27: 2427-2434. 198. Haruta M, Sasai Y, Kawasaki H, Amemiya K, Ooto S, et al. (2004) In vitro and in vivo characterization of pigment epithelial cells differentiated from primate embryonic stem cells. Invest Ophthalmol Vis Sci 45: 1020-1025. 199. Lu B, Malcuit C, Wang S, Girman S, Francis P, et al. (2009) Long-Term Safety and Function of RPE from Human Embryonic Stem Cells in Preclinical Models of Macular Degeneration. Stem Cells 27: 2126-2135. 200. LaVail MM (2001) Legacy of the RCS rat: impact of a seminal study on retinal cell biology and retinal degenerative diseases. Prog Brain Res 131: 617-627. 201. Wu W, Zeng Y, Li Z, Li Q, Xu H, et al. (2016) Features specific to retinal pigment epithelium cells derived from three-dimensional human embryonic stem cell cultures - a new donor for cell therapy. Oncotarget 7: 22819-22833. 202. Krohne TU, Westenskow PD, Kurihara T, Friedlander DF, Lehmann M, et al. (2012) Generation of Retinal Pigment Epithelial Cells from Small Molecules and OCT4 Reprogrammed Human Induced Pluripotent Stem Cells. Stem Cells Trans Med 1: 96-109.

205. Zou T, Gao L, Zeng Y, Li Q, Li Y, et al. (2019) Organoid-derived C-Kit/ SSEA4 human retinal progenitor cells promote a protective retinal microenvironment during transplantation in rodents. Nat Commun 10: 1205. 206. Schwartz SD, Hubschman J-P, Heilwell G, Franco-Cardenas V, Pan CK, et al. (2012) Embryonic stem cell trials for macular degeneration: a preliminary report. Lancet 379: 713-720. 207. Schwartz SD, Tan G, Hosseini H, Nagiel A (2016) Subretinal Transplantation of Embryonic Stem Cell-Derived Retinal Pigment Epithelium for the Treatment of Macular Degeneration: An Assessment at 4 Years. Invest Ophthalmol Vis Sci 57. 208. Schwartz SD, Regillo CD, Lam BL, Eliott D, Rosenfeld PJ, et al. (2015) Human embryonic stem cell-derived retinal pigment epithelium in patients with age-related macular degeneration and Stargardt’s macular dystrophy: follow-up of two open-label phase 1/2 studies. Lancet 385: 509-516. 209. Song WK, Park K-M, Kim H-J, Lee JH, Choi J, et al. (2015) Treatment of Macular Degeneration Using Embryonic Stem Cell-Derived Retinal Pigment Epithelium: Preliminary Results in Asian Patients. Stem Cell Reports 4: 860-872. 210. Diniz B, Thomas P, Thomas B, Ribeiro R, Hu Y, et al. (2013) Subretinal Implantation of Retinal Pigment Epithelial Cells Derived From Human Embryonic Stem Cells: Improved Survival When Implanted as a Monolayer. Invest Ophthalmol Vis Sci 54: 5087-5096. 211. Hsiung J, Zhu D, Hinton DR (2015) Polarized Human Embryonic Stem Cell-Derived Retinal Pigment Epithelial Cell Monolayers Have Higher Resistance to Oxidative Stress-Induced Cell Death Than Nonpolarized Cultures. Stem Cells Transl Med 4: 10-20. 212. White CE, Olabisi RM (2017) Scaffolds for retinal pigment epithelial cell transplantation in age-related macular degeneration. J Tissue Eng 8: 204173141772084. 213. da Cruz L, Fynes K, Georgiadis O, Kerby J, Luo YH, et al. (2018) Phase 1 clinical study of an embryonic stem cell-derived retinal pigment epithelium patch in age-related macular degeneration. Nat Biotechnol 2018, 36: 328-337. 214. Kashani AH, Lebkowski JS, Rahhal FM, Avery RL, Salehi-Had H, et al. (2018) A bioengineered retinal pigment epithelial monolayer for advanced, dry age-related macular degeneration. Sci Trans Med 10: 3. 215. Thomas BB, Zhu D, Zhang L, Thomas PB, Hu Y, et al. (2016) Survival and Functionality of hESC-Derived Retinal Pigment Epithelium Cells Cultured as a Monolayer on Polymer Substrates Transplanted in RCS Rats. Invest Ophthalmol Vis Sci 57: 2877-2887. 216. Koss MJ, Falabella P, Stefanini FR, Pfister M, Thomas BB, et al. (2016) Subretinal implantation of a monolayer of human embryonic stem cell-derived retinal pigment epithelium: a feasibility and safety study in Yucatan minipigs. Graefes Arch Clin Exp Ophthalmol 254: 1553-1565. 217. Brant Fernandes RA, Koss MJ, Falabella P, Stefanini FR, Maia M, et al. (2016) An Innovative Surgical Technique for Subretinal Transplantation of Human Embryonic Stem Cell-Derived Retinal Pigmented Epithelium in Yucatan Mini Pigs: Preliminary Results. Ophthalmic Surg Lasers Imaging Retina 47: 342-351. 218. Baker M (2013) Safety of induced stem cells gets a boost. Nature 493: 145-145.

203. Riera M, Fontrodona L, Albert S, Mora Ramirez D, Seriola A, et al. (2016) Comparative study of human embryonic stem cells (hESC) and human induced pluripotent stem cells (hiPSC) as a treatment for retinal dystrophies. Mol Ther Methods Clin Dev 3: 16010.

219. Araki R, Uda M, Hoki Y, Sunayama M, Nakamura M, et al. (2013) Negligible immunogenicity of terminally differentiated cells derived from induced pluripotent or embryonic stem cells Nature 494:100-104.

204. Sun JN, Mandai M, Kamao H, Hashiguchi T, Shikamura M, et al. (2015) Protective Effects of Human iPS-Derived Retinal Pigmented Epithelial Cells in Comparison with Human Mesenchymal Stromal Cells and Human Neural Stem Cells on the Degenerating Retina in rd1 Mice. Stem Cells 33: 1543-1553.

220. Kokkinaki M, Sahibzada N, Golestaneh N (2011) Human Induced Pluripotent Stem-Derived Retinal Pigment Epithelium (RPE) Cells Exhibit Ion Transport, Membrane Potential, Polarized Vascular Endothelial Growth Factor Secretion, and Gene Expression Pattern Similar to Native RPE. Stem Cells 29: 825-835.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 17 of 22 •

221. Yvon C, Ramsden CM, Lane A, Powner MB, da Cruz L, et al. (2015) Using Stem Cells to Model Diseases of the Outer Retina. Comput Struct Biotechnol J 13: 382-389. 222. Forest DL, Johnson LV, Clegg DO (2015) Cellular models and therapies for age-related macular degeneration. Dis Model Mech 8: 421-427. 223. Li Y, Tsai YT, Hsu CW, Erol D, et al (2012) Long-term Safety and Efficacy of Human-Induced Pluripotent Stem Cell (iPS) Grafts in a Preclinical Model of Retinitis Pigmentosa. Mol Med 18: 1312-1319. 224. Tucker BA, Park IH, Qi SD, Klassen HJ, Jiang C, et al. (2011) : Transplantation of Adult Mouse iPS Cell-Derived Photoreceptor Precursors Restores Retinal Structure and Function in Degenerative Mice. PloS One 6: 18992. 225. Hu Y, Liu L, Lu B, Zhu D, Ribeiro R, et al. (2012) A Novel Approach for Subretinal Implantation of Ultrathin Substrates Containing Stem Cell-Derived Retinal Pigment Epithelium Monolayer.Ophthalmic Res 48: 186-191. 226. Kamao H, Mandai M, Okamoto S, Sakai N, Suga A, et al. (2014) Characterization of Human Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium Cell Sheets Aiming for Clinical Application. Stem Cell Rep 2: 205-218. 227. Khristov V, Wan Q, Sharma R, Lotfi M, Maminishkis A, et al. (2018) Polarized Human Retinal Pigment Epithelium Exhibits Distinct Surface Proteome on Apical and Basal Plasma Membranes. Methods Mol Biol 1722: 223-247. 228. Sharma R, Khristov V, Rising A, Jha BS, Dejene R, et al. (2013) Clinical-grade stem cell-derived retinal pigment epithelium patch rescues retinal degeneration in rodents and pigs. Sci Transl Med 11. 229. Cyranoski D (2013) Stem cells cruise to clinic. Nature 494: 413-413. 230. Garber K (2015) RIKEN suspends first clinical trial involving induced pluripotent stem cells. Nat Biotechnol 33: 890-891. 231. Bartsch U, Oriyakhel W, Kenna PF, Linke S, Richard G, et al. (2008) Retinal cells integrate into the outer nuclear layer and differentiate into mature photoreceptors after subretinal transplantation into adult mice. Exp Eye Res 86: 691-700. 232. Radtke ND, Aramant RB, Petry HM, Green PT, Pidwell DJ, et al. (2008) Vision improvement in retinal degeneration patients by implantation of retina together with retinal pigment epithelium. Am J Ophthalmol 146: 172-182. 233. Pearson RA, Barber AC, Rizzi M, Hippert C, Xue T, et al. (2012) Restoration of vision after transplantation of photoreceptors. Nature 485: 99-103. 234. MacLaren RE, Pearson RA, MacNeil A, Douglas RH, Salt TE, et al. (2006) Retinal repair by transplantation of photoreceptor precursors. Nature 444: 203-207. 235. Calvert PD, Krasnoperova NV, Lyubarsky AL, Isayama T, Nicoló M, et al. (2000) Phototransduction in transgenic mice after targeted deletion of the rod transducin alpha-subunit. Proc Natl Acad Sci USA 97: 1391313918. 236. Gonzalez-Cordero A, West EL, Pearson RA, Duran Y, Carvalho LS, et al. (2013) Photoreceptor precursors derived from three-dimensional embryonic stem cell cultures integrate and mature within adult degenerate retina. Nat Biotechnol 31: 741-747. 237. Lakowski J, Gonzalez-Cordero A, West EL, Han Y-T, Welby E, etal. (2015) Transplantation of photoreceptor precursors isolated via a cell surface biomarker panel from embryonic stem cell-derived self-forming retina. Stem Cells 33: 2469-2482. J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

238. Waldron PV, Di Marco F, Kruczek K, Ribeiro J, Graca AB, et al. (2018) Transplanted donor- or stem cell-derived cone photoreceptors can both integrate and undergo material transfer in an environment-dependent manner. Stem Cell Rep 10: 406-421. 239. Mandai M, Fujii M, Hashiguchi T, Sunagawa GA, Ito S-i, et al. (2017) iPSC-derived retina transplants improve vision in rdl end-stage retinal-degeneration mice. Stem Cell Rep 8: 69-83. 240. Barber AC, Hippert C, Duran Y, West EL, Bainbridge JWB, et al. (2013) Repair of the degenerate retina by photoreceptor transplantation. Proc Natl Acad Sci U S A 110: 354-359. 241. Eberle D, Schubert S, Postel K, Corbeil D, Ader M (2011) Increased integration of transplanted CD73-positive photoreceptor precursors into adult mouse retina. Invest Ophthalmol Vis Sci 52: 6462-6471. 242. Lakowski J, Han YT, Pearson RA, Gonzalez-Cordero A, West EL, et al. (2011) Effective transplantation of photoreceptor precursor cells selected via cell surface antigen expression. Stem Cells 9: 1391-1404. 243. Kaplan HJ, Tezel TH, Berger AS, Wolf ML, Del Priore LV (1997) Human photoreceptor transplantation in retinitis pigmentosa. A safety study. Arch Ophthalmol 115: 1168-1172. 244. Radke ND, Aramant RB, Seiler MJ, Petry HM, Pidwell D (2004) Vision change after sheet transplant of fetal retina with retinal pigment epithelium to a patient with retinitis pigmentosa. Arch Ophthalmol 122: 11591165. 245. Berger AS, Tezel TH, Del Priore LV, Kaplan HJ (2003) Photoreceptor transplantation in retinitis pigmentosa: Short-term follow-up. Ophthalmology 110: 383-391. 246. Radtke ND, Aramant RB, Seiler M, Petry HM (1999) Preliminary report: Indications of improved visual function after retinal sheet transplantation in retinitis pigmentosa patients. Am J Ophthalmol 128: 384-387. 247. Hambright D, Park K-Y, Brooks M, McKay R, Swaroop A, et al. (2012) Long-term survival and differentiation of retinal neurons derived from human embryonic stem cell lines in un-immunosuppressed mouse retina. Mol Vis 18: 920-936. 248. Tsai Y, Lu B, Bakondi B, Girman S, Sahabian A, at al. (2015) Human iPSC-derived neural progenitors preserve vision in an AMD-like model. Stem Cells 33: 2537-2549. 249. Lu B, Morgans CW, Girman S, Luo J, Zhao J, at al. (2013) Neural stem cells derived by small molecules preserve vision. Trans Vis Sci Technolo1 2:1. 250. Luo J, Baranov P, Patel S, Ouyang H, Quach J, et al. (2014) Human retinal progenitor cell transplantation preserves vision. J Biol Chem 289: 6362-6371. 251. Furukawa T, Morrow EM, Cepko CL (1997) Crx, a novel otx-like homeobox gene, shows photoreceptor-specific expression and regulates photoreceptor differentiation. Cell 91: 531-541. 252. Tu H-Y, Watanabe T, Shirai H, Yamasaki S, Kinoshita M, et al. (2019) Medium- to long-term survival and functional examination of human iPSC-derived retinas in rat and primate models of retinal degeneration. EBioMedicine 39: 562-574. 253. Singh MS, Issa PC, Butler R, Martin C, Lipinski DM, et al. (2013) Reversal of end-stage retinal degeneration and restoration of visual function by photoreceptor transplantation. Proc Natl Acad Sci U S A 110: 11011106. 254. Smiley S, Nickerson PE, Comanita L, Daftarian N, El-Sehemy A, et al. (2016) Establishment of a cone photoreceptor transplantation platform based on a novel cone-GFP reporter mouse line. Sci Rep 6: 22867. 255. Santos-Ferreira T, Postel K, Stutzki H, Kurth T, Zeck G, et al. (2015) Daylight vision repair by cell transplantation. Stem Cells 33: 79-90.

Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 18 of 22 •

256. Lakowski J, Baron M, Bainbridge J, Barber AC, Pearson RA, et al., (2010) Cone and rod photoreceptor transplantation in models of the childhood retinopathy Leber congenital amaurosis using flow-sorted Crx-positive donor cells. Hum Mol Genet 19: 4545-4559.

274. Schlichtenbrede FC, da Cruz L, Stephens C, Smith AJ, Georgiadis A, Thrasher AJ, Bainbridge JWB, Seeliger MW, Ali RR (2003) Longterm evaluation of retinal function in Prph2(Rd2/Rd2) mice following AAV-mediated gene replacement therapy. J Gene Med 5: 757-764.

257. Pearson RA, Gonzalez-Cordero A, West EL, Ribeiro JR, Aghaizu N, et al., (2016) Donor and host photoreceptors engage in material transfer following transplantation of post-mitotic photoreceptor precursors. Nat Commun 7: 13029.

275. Ehlers JP, Petkovsek DS, Yuan A, Singh RP, Srivastava SK (2015) Intrasurgical Assessment of Subretinal tPA Injection for Submacular Hemorrhage in the PIONEER Study Utilizing Intraoperative OCT. Ophthalmic Surg Lasers Imaging Retina 46: 327-332.

258. Ortin Martinez A, Tsai ELS, Nickerson PE, Bergeret M, Lu Y, et al., (2017) A Reinterpretation of Cell Transplantation: GFP Transfer From Donor to Host Photoreceptors. Stem Cells 35: 932-939.

276. Price J, Turner D, Cepko C (1987) Lineage analysis in the vertebrate nervous system by retrovirus-mediated gene transfer. Proc Natl Acad Sci U S A 84:156-160.

259. Decembrini S, Martin C, Sennlaub F, Chemtob S, Bie M, et al., (2017) Cone Genesis Tracing by the Chrnb4-EGFP Mouse Line: Evidences of Cellular Material Fusion after Cone Precursor Transplantation. Mol Ther 25: 634-653.

277. Pfeffer B, Wiggert B, Lee L, Zonnenberg B, Newsome D, et al. (1983) The presence of a soluble interphotoreceptor retinol-binding protein (IRBP) in the retinal interphotoreceptor space. J Cell Physiol 117: 333341.

260. Santos Ferreira T, Llonch S, Borsch O, Postel K, Haas J, et al., (2016) Retinal transplantation of photoreceptors results in donor-host cytoplasmic exchange. Nat Commun 7: 13028.

278. Gerding H (2007) A new approach towards a minimal invasive retina implant. J Neural Eng 4: S30-S37.

261. Singla DK (2016) Stem cells and exosomes in cardiac repair. Curr Opin Pharmacol 27: 19-23.

279. Parikh S, Le A, Davenport J, Gorin MB, Nusinowitz S, et al. (2016) An Alternative and Validated Injection Method for Accessing the Subretinal Space via a Transcleral Posterior Approach. J Vis Exp 7.

262. Tannenbaum SE, Turetsky TT, Singer O, Aizenman E, Kirshberg S, et al., (2012) Derivation of Xeno-Free and GMP-Grade Human Embryonic Stem Cells-Platforms for Future Clinical Applications. PloS One 7: e35325.

280. Tomita M, Lavik E, Klassen H, Zahir T, Langer R, et al. (2005) Biodegradable polymer composite grafts promote the survival and differentiation of retinal progenitor cells. Stem Cells 23: 1579-1588.

263. Wiley LA, Burnight ER, DeLuca AP, Anfinson KR, Cranston CM, et al., (2016) cGMP production of patient-specific iPSCs and photoreceptor precursor cells to treat retinal degenerative blindness. Sci Rep 6: 30742.

281. Thomson RC, Giordano GG, Collier JH, Ishaug SL, Mikos AG, et al. (1996) Manufacture and characterization of poly(alpha-hydroxy ester) thin films as temporary substrates for retinal pigment epithelium cells. Biomaterials 17: 321-327.

264. Ramsden CM, Powner MB, Carr A JF, Smart MJK, da Cruz L, et al., (2013) Stem cells in retinal regeneration: past, present and future. Development 140: 2576-2585.

282. Weisz JM , Humayun MS , Juan ED, Del Cerro M, Sunness JS, et al. (1999) Allogenic fetal retinal pigment epithelial cell transplant in a patient with geographic atropy. Retina 19: 540-545.

265. Capowski EE, Samimi K, Mayerl SJ, Phillips MJ, Pinilla I, et al., (2019) Reproducibility and staging of 3D human retinal organoids across multiple pluripotent stem cell lines. Development 146.

283. Seiler MJ, Aramant RB (2012) Cell replacement and visual restoration by retinal sheet transplants. Prog Retin Eye Res 31: 661-687.

266. Ausayakhun S, Yuvaves P, Ngamtiphakom S, Prasitsilp J (2005) Treatment of cytomegalovirus retinitis in AIDS patients with intravitreal ganciclovir. J Med Assoc Thai 88 Suppl 9: S15-20.

284. Xiang P, Wu KC, Zhu Y, Xiang L, Li C, et al. (2014) A novel Bruch’s membrane-mimetic electrospun substrate scaffold for human retinal pigment epithelium cells. Biomaterials 35: 9777-9788.

267. Gaudana R, Jwala J, Boddu SHS, Mitra AK (2009) Recent Perspectives in Ocular Drug Delivery. Pharm Res 26: 1197-1216.

285. Jha BS, Bharti K (2015) Regenerating Retinal Pigment Epithelial Cells to Cure Blindness: A Road Towards Personalized Artificial Tissue. Curr Stem Cell Rep 1: 79-91.

268. Carr A JF, Smart MJK, Ramsden CM, Powner MB, da Cruz L, Coffey PJ (2013) Development of human embryonic stem cell therapies for age-related macular degeneration. Trends Neurosci 36: 385-395. 269. Timmers A, Zhang H, Squitieri A, Gonzalez Pola C (2001) Subretinal injections in rodent eyes: Effects on electrophysiology and histology of rat retina. Mol Vis 7: 131-137. 270. Qi Y, Dai X, Zhang H, He Y, Zhang Y, Han J, Zhu P, Zhang Y, Zheng Q, Li X, Zhao C, Pang J (2015) Trans-Corneal Subretinal Injection in Mice and Its Effect on the Function and Morphology of the Retina. PloS One 10: e0136523.

286. Ohno-Matsui K, Mori K, Ichinose S, Sato T, Wang J, et al. (2006) In vitro and in vivo characterization of iris pigment epithelial cells cultured on amniotic membranes. Mol Vis 12: 1022-1032. 287. Lee CJ, Fishman HA, Bent SF (2007) Spatial cues for the enhancement of retinal pigment epithelial cell function in potential transplants. Biomaterials 28: 2192-2201. 288. Tezel TH, Priore LVD, Kaplan HJ (2004) Reengineering of Aged Bruch’s Membrane to Enhance Retinal Pigment Epithelium Repopulation. Invest Ophthalmol Vis Sci 45: 3337-3348.

271. Johnson CJ, Berglin L, Chrenek MA, Redmond TM, Boatright JH, et al., (2008) Technical Brief: Subretinal injection and electroporation into adult mouse eyes. Mol Vis 14: 2211-2226.

289. Malafaya PB, Silva GA, Reis RL (2007) Natural-origin polymers as carriers and scaffolds for biomolecules and cell delivery in tissue engineering applications. Adv Drug Deliv Rev 59: 207-233.

272. Busskamp V, Duebel J, Balya D, Fradot M, Viney TJ, et al., (2010) Genetic Reactivation of Cone Photoreceptors Restores Visual Responses in Retinitis Pigmentosa. Science 329: 413-417.

290. Hynes SR, Lavik EB (2010) A tissue-engineered approach towards retinal repair: scaffolds for cell transplantation to the subretinal space. Graefes Arch Clin Exp Ophthalmol 248: 763-778.

273. Muhlfriedel R, Michalakis S, Garcia Garrido M, Biel M, Seeliger MW (2013) Optimized technique for subretinal injections in mice. Methods Mol Biol 935: 343-349.

291. Treharne AJ, Grossel MC, Lotery AJ, Thomson HA (2011) The chemistry of retinal transplantation: the influence of polymer scaffold properties on retinal cell adhesion and control. Br J Ophthalmol 95: 768-773.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 19 of 22 •

292. Gunatillake P, Mayadunne R, Adhikari R (2006) Recent developments in biodegradable synthetic polymers. Biotechnol Annu Rev 12: 301-347.

310. Silverman MS, Hughes SE (1989) Transplantation of photoreceptors to light-damaged retina. Invest Ophthalmol Vis Sci 30: 1684-1690.

293. Kundu J, Michaelson A, Baranov P, Young MJ, Carrier RL (2014) Approaches to cell delivery: substrates and scaffolds for cell therapy. Dev Ophthalmol 53: 143-154.

311. Radtke ND, Seiler MJ, Aramant RB, Petry HM, Ophthalmology DJ (2002) Transplantation of intact sheets of fetal neural retina with its retinal pigment epithelium in retinitis pigmentosa patients. Am J Ophthalmol 133: 544-550.

294. Pennington BO, Clegg DO (2016) Pluripotent Stem Cell-Based Therapies in Combination with Substrate for the Treatment of Age-Related Macular Degeneration. J Ocul Pharmacol Ther 32: 261-271.

312. Seiler MJ, Aramant RB (1998)Intact sheets of fetal retina transplanted to restore damaged rat retinas. Invest Ophthalmol Vis Sci 39: 2121-2131.

295. Worthington KS, Wiley LA, Guymon CA, Salem AK, Tucker BA (2016) Differentiation of Induced Pluripotent Stem Cells to Neural Retinal Precursor Cells on Porous Poly-Lactic-co-Glycolic Acid Scaffolds. J Ocul Pharmacol Ther 32: 310-316.

313. Shirai H, Mandai M, Matsushita K, Kuwahara A, Yonemura S, et al. (2016) Transplantation of human embryonic stem cell-derived retinal tissue in two primate models of retinal degeneration. Proc Natl Acad Sci U S A 113: 81-90.

296. Christiansen AT, Tao SL, Smith M, Wnek GE, Prause JU, et al. (2012) Subretinal implantation of electrospun, short nanowire, and smooth poly(ε-caprolactone) scaffolds to the subretinal space of porcine eyes. Stem Cells Int 454295.

314. McLelland BT, Lin B, Mathur A, Aramant RB, Thomas BB, et al. (2018) Transplanted hESC-Derived Retina Organoid Sheets Differentiate, Integrate, and Improve Visual Function in Retinal Degenerate Rats. Invest Ophthalmol Vis Sci 59: 2586-2603.

297. Thompson JR, Worthington KS, Green BJ, Mullin NK, Jiao C, et al. (2019) Two-photon polymerized poly(caprolactone) retinal cell delivery scaffolds and their systemic and retinal biocompatibility. Acta Biomater 94: 204-218.

315. West EL, Pearson RA, Barker SE, Luhmann UF, Maclaren RE, et al. (2010) Long-term survival of photoreceptors transplanted into the adult murine neural retina requires immune modulation. Stem Cells 28: 19972007.

298. Jayakody SA, Gonzalez-Cordero A, Ali RR, Pearson RA (2015) Cellular strategies for retinal repair by photoreceptor replacement. Prog Retin Eye Res 46: 31-66.

316. Xian B, Huang B (2015) The immune response of stem cells in subretinal transplantation. Stem Cell Res Ther 6: 161.

299. Kador KE, Goldberg JL (2012) Scaffolds and stem cells: delivery of cell transplants for retinal degenerations. Expert Rev Ophthalmol 7: 459-470. 300. Pearson RA (2014) Advances in repairing the degenerate retina by rod photoreceptor transplantation. Biotechnol Adv 32: 485-491. 301. Pearson RA, Hippert C, Graca AB, Barber AC (2014) Photoreceptor replacement therapy: challenges presented by the diseased recipient retinal environment. Vis Neurosci 31:333-344. 302. Warre-Cornish K, Barber AC, Sowden JC, Ali RR, Pearson RA (2014) Migration, integration and maturation of photoreceptor precursors following transplantation in the mouse retina. Stem Cells Dev 23:941-954. 303. Pearson RA, Barber AC, Rizzi M, Hippert C, Xue T, et al. (2012) Restoration of vision after transplantation of photoreceptors. Nature 485: 99-103. 304. Eberle D, Kurth T, Santos-Ferreira T, Wilson J, Corbeil D et.al (2012) Outer segment formation of transplanted photoreceptor precursor cells. PloS One 7. 305. Zhu J, Cifuentes H, Reynolds J, Lamba DA (2017) Immunosuppression via Loss of IL2rγ Enhances Long-Term Functional Integration of hESC-Derived Photoreceptors in the Mouse Retina. Cell Stem Cell 20: 374-384. 306. Ballios BG, Cooke MJ, Kooy Dvd, Biomaterials MS (2010) A hydrogel-based stem cell delivery system to treat retinal degenerative diseases. Biomaterials 31: 2555-2564.

317. Pritchard CD, Arnér KM, Langer RS, Ghosh FK (2010) Retinal transplantation using surface modified poly(glycerol-co-sebacic acid) membranes. Biomaterials 31: 7978-7984. 318. Redenti S, Neeley WL, Rompani S, Saigal S, Yang J, et al. (2009) Engineering retinal progenitor cell and scrollable poly (glycerol-sebacate) composites for expansion and subretinal transplantation. Biomaterials 30: 3405-3414. 319. Tao S, Young C, Redenti S, Zhang Y, Klassen H, et al. (2007) Survival, migration and differentiation of retinal progenitor cells transplanted on micro-machined poly(methyl methacrylate) scaffolds to the subretinal space. Lab Chip 7: 695-701. 320. Neeley WL, Redenti S, Klassen H, Tao S, Desai T, et al. (2008) A microfabricated scaffold for retinal progenitor cell grafting. Biomaterials 29: 418-426. 321. Tezcaner A, Hicks D (2008) In vitro characterization of micropatterned PLGA-PHBV8 blend films as temporary scaffolds for photoreceptor cells. J Biomed Mater Res A 86: 170-181. 322. Yao J, Ko CW, Baranov PY, Regatieri CV, Redenti S, et al. (2015) Enhanced differentiation and delivery of mouse retinal progenitor cells using a micropatterned biodegradable thin-film polycaprolactone scaffold. Tissue Eng Part A 21: 1247-1260. 323. Jung YH, Phillips MJ, Lee J, Xie R, Ludwig AL, et al. (2018) 3D microstructured scaffolds to support photoreceptor polarization and maturation. Adv Mater Weinheim 30: 1803550.

307. Steedman MR, Tao SL, Klassen H, Microdevices TA (2010) Enhanced differentiation of retinal progenitor cells using microfabricated topographical cues. Biomed Microdevices 12: 363-369.

324. Stanzel BV, Liu Z, Somboonthanakij S, Wongsawad W, Brinken R, et al. (2014) Human RPE stem cells grown into polarized RPE monolayers on a polyester matrix are maintained after grafting into rabbit subretinal space. Stem Cell Rep 2: 64-77.

308. Tucker BA, Redenti SM, Jiang C, Swift JS, Klassen HJ, et al. (2010)The use of progenitor cell/biodegradable MMP2–PLGA polymer constructs to enhance cellular integration and retinal repopulation. Biomaterials 31: 9-19.

325. Li Y, Wu W-H, Hsu C-W, Nguyen HV, Tsai Y-T, et al. (2014) Gene therapy in patient-specific stem cell lines and a preclinical model of retinitis pigmentosa with membrane frizzled-related protein defects. Mol Ther 22: 1688-1697.

309. Redenti S, Tao S, Yang J, Gu P, Klassen H, et al. (2008) Retinal tissue engineering using mouse retinal progenitor cells and a novel biodegradable, thin-film poly(e-caprolactone) nanowire scaffold. J Ocul Biol Dis Infor 1:19-29.

326. Mehat MS (2016) Phase I/II clinical trial of human embryonic stem cell (hESC)-derived retinal pigmented epithelium (RPE) transplantation in Stargardt disease (STGD): One-year results. Invest Ophthalmol Vis Sci 57.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 20 of 22 •

327. Kurimoto Y, Hirami Y, Fujihara M, Morinaga C, Yamamoto M, et al. (2016) Transplantation of autologous induced pluripotent stem cell-derived retinal pigment epithelium cell sheets for exudative age related macular degeneration: A Pilot Clinical Study. Invest Ophthalmol Vis Sci 57. 328. Schwartz SD, Tan G, Hosseini H, Nagiel A (2016) Subretinal transplantation of embryonic stem cell-derived retinal pigment epithelium for the treatment of macular degeneration: An assessment at 4 years. Invest Ophthalmol Vis Sci 57: 1-9. 329. Parker J, Mitrousis N, Shoichet MS (2016) Hydrogel for simultaneous tunable growth factor delivery and enhanced viability of encapsulated cells in vitro. Biomacromolecules 17: 476-484. 330. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, et al. (2007) Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol 2: 751. 331. Miyagishima KJ, Wan Q, Corneo B, Sharma R, Lotfi MR, et al. (2016) In pursuit of authenticity: Induced pluripotent stem cell-derived retinal pigment epithelium for clinical applications. Stem Cells Transl Med 5: 1562-1574. 332. Morizane A, Kikuchi T, Hayashi T, Mizuma H, Takara S, et al. (2017) MHC matching improves engraftment of iPSC-derived neurons in non-human primates. Nat Commun 8: 385. 333. Sugita S, Iwasaki Y, Makabe K, Kimura T, Futagami T, et al. (2016) Lack of T cell response to iPSC-derived retinal pigment epithelial cells from HLA homozygous donors. Stem Cell Reports 7: 619-634. 334. De Rham C, Villard J (2014) Potential and limitation of HLA-based banking of human pluripotent stem cells for cell therapy. J Immunol Res 2014: 518135. 335. Gornalusse GG, Hirata RK, Funk SE, Riolobos L, Lopes VS, et al. (2017) HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells. Nat Biotechnol 2017 35: 765-772. 336. Eberle D, Santos-Ferreira T, Grahl S, Ader M (2014) Subretinal Transplantation of MACS Purified Photoreceptor Precursor Cells into the Adult Mouse Retina. J Vis Exp 84: 50932. 337. Thomas ED Sr (1997) Stem cell transplantation: past, present and future. Arch Immunol Ther Exp 45: 1-5. 338. Lakowski J, Welby E, Budinger D, Di Marco F, et al. (2018) Isolation of Human Photoreceptor Precursors via a Cell Surface Marker Panel from Stem Cell-Derived Retinal Organoids and Fetal Retinae. Stem Cells 36: 709-722. 339. Gagliardi G, Ben M’Barek K, Chaffiol A, Slembrouck-Brec A, Conart JB, et al. (2018) Characterization and Transplantation of CD73-Positive Photoreceptors Isolated from Human iPSC-Derived Retinal Organoids. Stem Cell Rep 11: 665-680. 340. Lidgerwood GE, Morris AJ, Conquest A, Daniszewski M, Rooney LA, et al. (20108) Role of lysophosphatidic acid in the retinal pigment epithelium and photoreceptors. Biochim Biophys Acta Mol Cell Biol Lipids 1863: 750-761. 341. Yao J, Tao SL, Young MJ (2011) Synthetic Polymer Scaffolds for Stem Cell Transplantation in Retinal Tissue Engineering. Polymers 3: 899914. 342. Nickerson PEB, Ortin-Martinez A, Wallace VA (2018) Material Exchange in Photoreceptor Transplantation: Updating Our Understanding of Donor/Host Communication and the Future of Cell Engraftment Science. Front Neural Circuits 12: 17. 343. Waldron PV, Di Marco F, Kruczek K, Ribeiro J, Graca AB, et al. (2018) Transplanted Donor- or Stem Cell-Derived Cone Photoreceptors Can Both Integrate and Undergo Material Transfer in an Environment-Dependent Manner. Stem Cell Rep 10: 406-421.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

344. Wu H, Li J, Mao X, Li G, Xie L, et al. (2018) Transplantation of rat embryonic stem cell-derived retinal cells restores visual function in the Royal College of Surgeons rats. Doc Ophthalmol 137: 71-78. 345. Collin J, Zerti D, Queen R, Santos-Ferreira T, Bauer R, et al. (2019) CRX Expression in Pluripotent Stem Cell-Derived Photoreceptors Marks a Transplantable Subpopulation of Early Cones. Stem Cells 37: 609-622. 346. Giacalone JC, Wiley LA, Burnight ER, Songstad AE, Mullins RF, et al. (2016) Concise Review: Patient-Specific Stem Cells to Interrogate Inherited Eye Disease. Stem Cells Transl Med 5: 132-140. 347. Phillips MJ, Capowski EE, Petersen A, Jansen AD, Barlow K, et al. (2018)Generation of a rod-specific NRL reporter line in human pluripotent stem cells. Sci Rep 8: 2370. 348. Wei W, Wang ZD, Zheng XL, Ding L, Han DM, et al. (2017)Clinical Observation of Haploidentical Hematopoietic Stem Cell Transplantation Combined with Bone-marrow Derived Mesenchymal Stem Cells Transfusion for Treatment of Children with Severe Aplastic Anemia. Zhongguo shi yan xue ye xue za zhi 25: 1158-1164. 349. Maklakova IY, Grebnev DY (2017) Effects of Combined Transplantation of Multipotent Mesenchymal Stromal and Hemopoietic Stem Cells on Regeneration of the Hemopoietic Tissue. Bull Exp Biol Med 163: 61-64. 350. Shujie W (2017) Effect of CB-EPCs Combined with Hematopoietic Stem Cell Transplantation on Hematopoietic Reconstitution and GVHD. Genomics & Applied Biology 2017. 351. Souza LC, Carvalho KA, Rebelatto C, Senegaglia A, Furuta M, et al. (2004) Combined transplantation of skeletal myoblasts and mesenchymal cells (cocultivation) in ventricular dysfunction after myocardial infarction.Arq Bras Cardiol 83: 294-299; 288-293. 352. Ohmura Y, Tanemura M, Kawaguchi N, Machida T, Tanida T, et al. (2010)Combined transplantation of pancreatic islets and adipose tissue-derived stem cells enhances the survival and insulin function of islet grafts in diabetic mice. Transplantation 90: 1366-1373. 353. Fang JF, Huang XN, Han XY, Ouyang X, Fan L, et al. (2018)Combined Transplantation of Mesenchymal Stem Cells and Endothelial Progenitor Cells Restores Cavernous Nerve Injury-Related Erectile Dysfunction. J Sex Med 15: 284-295. 354. Johnson TV, Martin KR (2013) Cell transplantation approaches to retinal ganglion cell neuroprotection in glaucoma. Curr Opin Pharmacol 13: 78-82. 355. Pearson C, Martin K (2015) Stem cell approaches to glaucoma: from aqueous outflow modulation to retinal neuroprotection. Prog Brain Res 220: 241-256. 356. Mao X, Pan T, Shen H, Xi H, Yuan S, et al. (2018) The rescue effect of mesenchymal stem cell on sodium iodate-induced retinal pigment epithelial cell death through deactivation of NF-κB-mediated NLRP3 inflammasome. Biomed Pharmacother 103: 517-523. 357. Qu L, Gao L, Xu H, Duan P, Zeng Y, et al. (2017) Combined transplantation of human mesenchymal stem cells and human retinal progenitor cells into the subretinal space of RCS rats. Sci Rep 7: 199. 358. Wu S, Chang KC, Nahmou M, Goldberg JL (2018) Induced Pluripotent Stem Cells Promote Retinal Ganglion Cell Survival After Transplant. Invest Ophthalmol Vis Sci 59: 1571-1576. 359. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, et al. (2010) Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330: 841-845. 360. Tambuyzer BR, Ponsaerts P, Nouwen EJ (2009) Microglia: gatekeepers of central nervous system immunology. J Leukoc Biol 85: 352-370. Volume 5 • Issue 1 • 100014


Citation: Zhang C-J, Jin Z-B (2019) Restoring Vision with Photoreceptor Regeneration. Stem Cell Res Dev Ther 4: 014.

• Page 21 of 22 •

361. Neumann J, Sauerzweig S, Rönicke R, Gunzer F, Dinkel K, et al. (2008) Microglia cells protect neurons by direct engulfment of invading neutrophil granulocytes: a new mechanism of CNS immune privilege. J Neurosci 28: 5965-5975.

372. Gonzalez-Cordero A, West EL, Pearson RA, Duran Y, Carvalho LS, et al. (2013) Photoreceptor precursors derived from three-dimensional embryonic stem cell cultures integrate and mature within adult degenerate retina. Nat Biotechnol 31: 741-747.

362. Kawabori M, Kacimi R, Kauppinen T, Calosing C, Kim JY, et al. (2015) Triggering receptor expressed on myeloid cells 2 (TREM2) deficiency attenuates phagocytic activities of microglia and exacerbates ischemic damage in experimental stroke. J Neurosci 35: 3384-3396.

373. Lewin AS, Rossmiller B, Mao H (2014) Gene augmentation for adRP mutations in RHO. Cold Spring Harb Perspect Med 4: 017400.

363. Okunuki Y, Mukai R, Pearsall EA, Klokman G, Husain D, et al. (2018) Microglia inhibit photoreceptor cell death and regulate immune cell infiltration in response to retinal detachment. Proc Natl Acad Sci USA 115: 6264-6273. 364. O’Koren EG, Yu C, Klingeborn M, Wong AYW, Prigge CL, et al. (2019) Microglial Function Is Distinct in Different Anatomical Locations during Retinal Homeostasis and Degeneration. Immunity 50: 723-737. 365. Muffat J, Li Y, Yuan B, Mitalipova M, Omer A, et al. (2016) Efficient derivation of microglia-like cells from human pluripotent stem cells. Nat Med 22: 1358-1367. 366. Pandya H, Shen MJ, Ichikawa DM, Sedlock AB, Choi Y, et al (2017) Differentiation of human and murine induced pluripotent stem cells to microglia-like cells. Nat Neurosci 20: 753-759. 367. Sohocki MM, Daiger SP, Bowne SJ, Rodriquez JA, Northrup H, et al. (2001) Prevalence of mutations causing retinitis pigmentosa and other inherited retinopathies. Hum Mut 17: 42-51. 368. D’Cruz PM, Yasumura D, Weir J, Matthes MT, Abderrahim H, et al. (2000) Mutation of the receptor tyrosine kinase gene Mertk in the retinal dystrophic RCS rat. Hum Mol Genet 9: 645-651. 369. Baranov PY, Tucker BA, Young MJ (2014) Low-Oxygen Culture Conditions Extend the Multipotent Properties of Human Retinal Progenitor Cells. Tissue Eng Part A 20: 1465-1475. 370. Semo M, Haamedi N, Stevanato L, Carter D, Brooke G, et al. (2016) Efficacy and Safety of Human Retinal Progenitor Cells. Trans Vis Sci Technol 5: 6. 371. Haruta M, Bush RA, Kjellstrom S, Vijayasarathy C, Zeng Y, et al. (2009) Depleting Rac1 in mouse rod photoreceptors protects them from photo-oxidative stress without affecting their structure or function. Proc Natl Acad Sci USA 106: 9397-9402.

J Stem Cell Res Dev Ther ISSN: 2381-2060, Open Access Journal DOI: 10.24966/SRDT-2060/100014

374. Reuter JH, Sanyal SJ (1984) Development and degeneration of retina in rds mutant mice: The electroretinogram. Neurosci Lett 48: 231-237. 375. Lolley RN, Rong H, Craft CM (1994) Linkage of photoreceptor degeneration by apoptosis with inherited defect in phototransduction. Invest Ophthalmol Vis Sci 35: 358-362. 376. Mears AJ, Kondo M, Swain PK, Takada Y, Bush RA, et al. (2001) Nrl is required for rod photoreceptor development. Nat Genet 29: 447-452. 377. Samardzija M, Caprara C, Heynen SR, Willcox DeParis S, Meneau I, et al. (2014) A mouse model for studying cone photoreceptor pathologies. Invest Ophthalmol Vis Sci 55: 5304-5313. 378. Lamba DA, Karl MO, Ware CB, Reh TA (2006) Efficient generation of retinal progenitor cells from human embryonic stem cells. Proc Natl Acad Sci USA 103: 12769-12774. 379. Furukawa T, Morrow EM, Li T, Davis FC, Cepko CL (1999) Retinopathy and attenuated circadian entrainment in Crx-deficient mice. Nat Genet 23: 466-470. 380. Martinez-Navarrete G, Seiler MJ, Aramant RB, Fernandez-Sanchez L, Pinilla I, et al. (2011) Retinal degeneration in two lines of transgenic S334ter rats. Exp Eye Res 92:227-237. 381. McLelland BT, Lin B, Mathur A, Aramant RB, Thomas BB, et al. (2018) Transplanted hESC-Derived Retina Organoid Sheets Differentiate, Integrate, and Improve Visual Function in Retinal Degenerate Rats. Invest Ophthalmol Vis Sci 59: 2586-2603. 382. Iraha S, Tu HY, Yamasaki S, Kagawa T, Goto M, et al. (2018) Establishment of Immunodeficient Retinal Degeneration Model Mice and Functional Maturation of Human ESC-Derived Retinal Sheets after Transplantation. Stem Cell Rep 10: 1059-1074. 383. Seiler MJ, Aramant RB, Jones MK, Ferguson DL, Bryda EC, et al. (2014) A new immunodeficient pigmented retinal degenerate rat strain to study transplantation of human cells without immunosuppression. Graefes Arch Clin Exp Ophthalmol 252: 1079-1092.

Volume 5 • Issue 1 • 100014


Journal of Anesthesia & Clinical Care

Journal of Gerontology & Geriatric Medicine

Journal of Addiction & Addictive Disorders

Journal of Genetics & Genomic Sciences

Advances in Microbiology Research

Journal of Hematology, Blood Transfusion & Disorders

Advances in Industrial Biotechnology

Journal of Human Endocrinology

Journal of Agronomy & Agricultural Science

Journal of Hospice & Palliative Medical Care

Journal of AIDS Clinical Research & STDs

Journal of Internal Medicine & Primary Healthcare

Journal of Alcoholism, Drug Abuse & Substance Dependence

Journal of Infectious & Non Infectious Diseases

Journal of Allergy Disorders & Therapy

Journal of Light & Laser: Current Trends

Journal of Alternative, Complementary & Integrative Medicine

Journal of Modern Chemical Sciences

Journal of Alzheimer’s & Neurodegenerative Diseases

Journal of Medicine: Study & Research

Journal of Angiology & Vascular Surgery

Journal of Nanotechnology: Nanomedicine & Nanobiotechnology

Journal of Animal Research & Veterinary Science

Journal of Neonatology & Clinical Pediatrics

Archives of Zoological Studies

Journal of Nephrology & Renal Therapy

Archives of Urology

Journal of Non Invasive Vascular Investigation

Journal of Atmospheric & Earth-Sciences

Journal of Nuclear Medicine, Radiology & Radiation Therapy

Journal of Aquaculture & Fisheries

Journal of Obesity & Weight Loss

Journal of Biotech Research & Biochemistry

Journal of Ophthalmology & Clinical Research

Journal of Brain & Neuroscience Research

Journal of Orthopedic Research & Physiotherapy

Journal of Cancer Biology & Treatment

Journal of Otolaryngology, Head & Neck Surgery

Journal of Cardiology: Study & Research

Journal of Protein Research & Bioinformatics

Journal of Cell Biology & Cell Metabolism

Journal of Pathology Clinical & Medical Research

Journal of Clinical Dermatology & Therapy

Journal of Pharmacology, Pharmaceutics & Pharmacovigilance

Journal of Clinical Immunology & Immunotherapy

Journal of Physical Medicine, Rehabilitation & Disabilities

Journal of Clinical Studies & Medical Case Reports

Journal of Plant Science: Current Research

Journal of Community Medicine & Public Health Care

Journal of Psychiatry, Depression & Anxiety

Current Trends: Medical & Biological Engineering

Journal of Pulmonary Medicine & Respiratory Research

Journal of Cytology & Tissue Biology

Journal of Practical & Professional Nursing

Journal of Dentistry: Oral Health & Cosmesis

Journal of Reproductive Medicine, Gynaecology & Obstetrics

Journal of Diabetes & Metabolic Disorders

Journal of Stem Cells Research, Development & Therapy

Journal of Dairy Research & Technology

Journal of Surgery: Current Trends & Innovations

Journal of Emergency Medicine Trauma & Surgical Care

Journal of Toxicology: Current Research

Journal of Environmental Science: Current Research

Journal of Translational Science and Research

Journal of Food Science & Nutrition

Trends in Anatomy & Physiology

Journal of Forensic, Legal & Investigative Sciences

Journal of Vaccines Research & Vaccination

Journal of Gastroenterology & Hepatology Research

Journal of Virology & Antivirals

Submit Your Manuscript: http://www.heraldopenaccess.us/Online-Submission.php Herald Scholarly Open Access, 2561 Cornelia Rd, #205, Herndon, VA 20171, USA. Tel: +1 202-499-9679; E-mail: info@heraldsopenaccess.us http://www.heraldopenaccess.us/


Turn static files into dynamic content formats.

Create a flipbook
Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.