Skip to main content
  • Review
  • Published:

Ocular stem cells: a status update!

Abstract

Stem cells are unspecialized cells that have been a major focus of the field of regenerative medicine, opening new frontiers and regarded as the future of medicine. The ophthalmology branch of the medical sciences was the first to directly benefit from stem cells for regenerative treatment. The success stories of regenerative medicine in ophthalmology can be attributed to its accessibility, ease of follow-up and the eye being an immune-privileged organ. Cell-based therapies using stem cells from the ciliary body, iris and sclera are still in animal experimental stages but show potential for replacing degenerated photoreceptors. Limbal, corneal and conjunctival stem cells are still limited for use only for surface reconstruction, although they might have potential beyond this. Iris pigment epithelial, ciliary body epithelial and choroidal epithelial stem cells in laboratory studies have shown some promise for retinal or neural tissue replacement. Trabecular meshwork, orbital and sclera stem cells have properties identical to cells of mesenchymal origin but their potential has yet to be experimentally determined and validated. Retinal and retinal pigment epithelium stem cells remain the most sought out stem cells for curing retinal degenerative disorders, although treatments using them have resulted in variable outcomes. The functional aspects of the therapeutic application of lenticular stem cells are not known and need further attention. Recently, embryonic stem cell-derived retinal pigment epithelium has been used for treating patients with Stargardts disease and age-related macular degeneration. Overall, the different stem cells residing in different components of the eye have shown some success in clinical and animal studies in the field of regenerative medicine.

Introduction

Pluripotency, the capacity to differentiate into multiple lineages, and proliferation are two characteristic attributes of stem cells. These cells are capable of replacing damaged or diseased cells under certain circumstances. Regenerative medicine or stem cell-based therapy has now reached a state where ocular tissues damaged by disease or injury can be repaired and/or regenerated. The ease of access for the therapeutic procedure as well as follow-up together with its immune-privileged status makes the eye an ideal organ for studying regenerative medicine. Such therapy involves various procedures where stem cells are injected into both the cellular and extracellular matrix microenvironments [1]. Corneal epithelial cell transplantation has been the most widely used stem cell-based therapy following bone marrow transplantation.

Stem cell-based treatment in ophthalmology follows either a cell replacement therapy strategy or a strategy involving trophic factor-based guidance cues. Throughout treatment, outcomes depend on our in-depth knowledge of the disease, the source of stem cells, the mode of treatment and the plausible mechanism driving the therapeutic outcome [2].

In this review we discuss region-specific stem cell populations and their respective functions in cell-based therapy. We also address possible hurdles to therapy and means to overcome these in our pursuit of regenerative medicine applications in the field of ophthalmology.

Cornea (limbus and stroma)

The cornea is at the outermost surface of the eye and safeguards transparency, which is crucial for vision. The corneal stem cell population is located in the periphery of the cornea, in the limbus; these cells are termed limbal epithelial stem cells (LESCs) [3–6]. Stroma comprises 90% of the volume of the cornea and, unlike the self-renewal of epithelia, the homeostasis of stroma is not based on a cycle of cell death and mitotic renewal.

Identification and isolation

Stem cells in the corneal epithelium are located in the basal layer of the limbal region at the corneal periphery, called the palisades of Vogt [3]. These are visualized in small clusters and are closely associated with the stromal matrix and the basement membrane, thereby assisting in cell-cell, cell-extracellular matrix and paracrine signaling communication. The corneal epithelial basal layer is composed mostly of transient amplifying cells at various stages of maturity.

LESCs are identified by their elevated expression of an isoform of the transcription factor p63 along with a high nuclear to cytoplasmic ratio [7, 8]. ABCG2 (ATP binding cassette sub family G member 2) positivity has been detected in LESCs as well as several other cells residing in the suprabasal limbus and these markers have the potential to identify the LESC population based on their staining ability in clusters of progenitor-like cells in the limbus [9, 10]. Reports also indicate that Musashi-1, an RNA binding protein, can be used to specifically stain LESCs [11, 12]. Corneal stem cells also express enolase, cytokeratin (CK)19, and vimentin but do not express CK3, CK12, or Connexin 43, which are present in corneal epithelial cells [11, 12].

Stromal multipotent clonal cells have been identified and expanded to neurospheres in cultures [13, 14]. Corneal stromal stem cells are located in the anterior stroma sub-adjacent to the basal side of the palisades of Vogt [15]. Stem cells in the stroma were identified as a side population using the DNA-binding dye Hoechst 33342. These cells expressed genes encoding ABCG2, Bmi1, CD166, c-kit, Pax6, Six2 and Notch1 as well as mesenchymal stem cell and early corneal developmental markers. When differentiated, corneal stromal stem cells expressed keratocyte markers such as keratocan, ALDH3A1, CXADR, PTDGS and PDK4 [16].

Therapeutic implications

LESC deficiency is pathological, either partially or completely, and is caused by either mechanical injury or chemical and thermal burns or acquired by diseases such as aniridia and Stevens Johnson syndrome. Treatment of such conditions involves LESC transplantation therapy. LESCs from the healthy eye in unilateral cases of ocular disease are expanded ex vivo for therapeutic purposes using protocols involving amniotic membrane or fibrin in the presence or absence of growth-arrested 3 T3 fibroblast feeder layers. Alternative, experimental sources for LESCs for cell-based therapy include buccal mucosal epithelial cells, hair follicle stem cells, and human embryonic stem cells (ESCs) [17, 18]. Among non-limbal cell types, cultured oral mucosal cells and conjunctival epithelial cells have been transplanted to treat limbal stem cell deficiency in humans [19, 20].

Recent research shows that the peripheral cornea contains a higher density of keratocyte precursors with high proliferative capacity. A three-dimensional construction using corneal keratocyte precursors and gelatin hydrogels provided cues for attracting keratocytes and extracellular matrix in scarred stroma [21]. Du and colleagues [22] demonstrated restoration of corneal transparency, stromal thickness and collagen fibril defects after injecting corneal stromal stem cells in mice. If successful, such therapy would eliminate the shortage of donor corneas needed for transplantations. Although stem cell transplantation is performed worldwide, variability in clinical outcomes implies that standardized protocols need to be established. Further validation and quality assessment studies on these cell types could provide therapeutic solutions for ocular surface reconstruction, and may also provide insights into the feasibility of their use for reconstruction of tissues beyond the ocular surface.

Conjunctiva

The conjunctiva, apart from being a barrier to pathogenic entry, is a highly vascularized connective tissue that provides channels for proper flow of nutrients and fluids. Conjunctival cells undergo renewal similar to the corneal epithelium, but the source of the stem cells for this remains elusive [23].

Identification and isolation

Conjunctival stem cells can differentiate into either mucin-producing goblet cells or an epithelial cell. The dividing basal cells migrate from the bulbar conjunctiva to the corneal surface and differentiate. Conjunctival epithelial cells are negative for CK3 and CK12 but positive for CK19. The stem cells residing in the fornical niche can differentiate into epithelial cells as well as goblet cells, as shown in clonal culture assays. This provides strong evidence that the stem cell population for conjunctiva renewal is in the fornix region [24, 25].

Therapeutic implications

Ocular processes that affect the cornea also affect the conjunctiva. Conjunctival scarring, cicatricial pemphigoid, thickening, dry eye or mucin deficiency are some of the conditions affecting the conjunctiva. Conjunctival autografts, oral mucous membrane grafts, nasal turbinate mucosa grafts and amniotic membrane are often used to treat conjunctival stem cell deficiency and scarring [18]. Conjunctival cells cultured on amniotic membrane have been used for cell transplantation in patients with limbal stem cell deficiency. Recent patient follow-up reports have shown that transplantation of autologous conjunctival epithelial cells improved the clinical parameters of total limbal stem cell deficiency with respect to vision acuity, impression cytology and in vivo confocal analysis [18, 26]. These cells were cultivated ex vivo (on amniotic membrane) in Dulbecco's modified Eagle's medium with Ham’s 12 in the presence of epidermal growth factor, insulin, cholera toxin and hydrocortisone to derive the corneal lineage; the cells were transplanted after 2 weeks of culture. Ultrathin polymembrane (epsilon-caprolactone) substrate has also been shown to support conjunctival epithelial cell proliferation [27].

Iris

The iris divides the space between the cornea and lens into anterior and posterior halves. The stroma and the vasculature of the iris are developed from the anterior region of the optic cup [28].

Identification and isolation

Iris pigment epithelial cells have the ability to grow in spheres and express markers of neural stem/progenitor cells such as Nestin, Msi and Pax6. Studies from mouse iris have revealed that these cells can also be differentiated to neuronal as well as glial lineages and express markers such as Chx10, Rho, Otx2 and Olig2 [29].

Therapeutic implications

Though the iris pigment epithelial cells have potential to be used in cell-based therapy, not much work on validation and quality assessment has been done. These cells can be transdifferentiated into retinal neuronal cells expressing retinal-specific markers [30]. Further studies are needed before iris pigment epithelial cells can be used clinically.

Ciliary body

The ciliary body produces the aqueous humor and is involved in regulating the aqueous flow, blood flow, intra-ocular pressure and maintenance of the immune-privileged status of the anterior chamber [31].

Identification and isolation

Ciliary body stem cells are derived from ciliary epithelium and undergo lineage-specific differentiation to retinal tissues. The ciliary-derived progenitor cell population expresses neuronal/retinal markers such as Nestin, Chx10 and Pax6. Ciliary epithelial cells can be cultured in vitro, forming neurospheres expressing transcription factors (Sox 2 and Pax 6) and retinal markers (Lhx2, Dach1, Six 3) [32].

Therapeutic implications

During homeostasis ciliary epithelium maintains a balance between epithelial and neuronal cell types, whereas during disease ciliary epithelium cells can act as donor cells for retinal repair. Studies so far have revealed that ciliary epithelium cells differentiate well into the retinal lineage cells that express retinal markers but do not integrate with existing retinal architecture. Recently, Gualdoni and colleagues [33] and Yanagi and colleagues [34] reported that ciliary epithelium cells lack the potential to differentiate into photoreceptors, suggesting that the cells need to be reprogrammed to be useful as a source of new photoreceptors. Further studies are warranted so we might realize the potential of these cells in clinics. Cicero and colleagues [35] reported that, although ciliary epithelium stem cells expressed retinal markers, each cell contained pigments and had membrane interdigitations and epithelial junctions. Ballios and colleagues [36] showed that clonally derived retinal stem cell progeny from ciliary epithelium can differentiate into mature rhodopsin-positive cells using a combination of exogenous culture additives (fibroblast growth factor, heparin, retionic acid, taurine). Inoue and colleagues [37] demonstrated that modulation of the retinal transcriptional factors OTX2, CRX and CHX10 increases the potential of retinal stem cell progeny derived from the cilliary margin of adult human eye.

Trabecular meshwork

The trabecular meshwork (TM) is a tissue between the cornea and iris in the anterior region that is responsible for drainage of aqueous fluid. The balance between aqueous secretion and outflow determines intraocular pressure, which is a risk factor for the development of glaucoma. TM cells help to remove debris in the circulating aqueous humor [38].

Identification and isolation

TM cells express vimentin, non-muscle actin, aquaporin-1, acetylated and acetoacetylated alpha-2 adrenergic receptor, matrix GLA protein and chitinase-3-like-1 [39–41]. Recently, the isolation and characterization of TM cells have been widely studied. These studies suggest that TM cells have stem cell-like properties, expressing mesenchymal cell-associated markers such as CD73, CD90, and CD105, and the ability to differentiate into adipocytes, osteocytes, and chondrocytes [38, 42]. Further, studies showed that TM stem cells isolated as a side population or as clones expressed specific stem cell markers such as ABCG2, Notch1, OCT-3/4, AnkG, and MUC1 [38]. These stem cells could differentiate into the TM lineage and expressed AQP1, CHI3L1, and TIMP3 markers and had a phagocytic function [38, 42].

Therapeutic implications

Lowering the intra-ocular pressure is an aim of treatments for glaucoma. The idea for this came primarily from the observation that TM cell division increased after argon laser trabeculoplasty [43]. Topical and oral medications, argon laser trabeculoplasty and some surgical approaches (for example, implant blebs) are current first-line treatments. A very recent study reported that stem cells isolated from human TM and expanded in vitro showed evidence of the ability to home to mouse TM and differentiate into TM cells in vivo[44]. The expanded TM stem cells expressed the stem cell markers ABCG2, Notch1, and MUC1 and were positive for expression of the TM marker protein CHI3L1. These TM cells were multipotent and had phagocytic properties [38, 45]. Some groups are working on transplanting TM cells or TM progenitor cells combined with argon laser trabeculoplasty as a novel cell-based therapy for glaucoma [38, 43–45].

Lens

The lens is composed of the lens capsule, epithelium and fibers and, like the cornea, is transparent. Lens stem cells are hypothesized to reside in the lens capsule, although they have not yet been identified. It is plausible that they come from the ciliary body, which is anatomically close to the lens [46].

Identification and isolation

Lens capsule regeneration has been shown to occur in lower vertebrates from cells residing in the ciliary body. The lens stem cells might thus reside in the lens capsule [47, 48]. Lens stem cells have not yet been identified.

Therapeutic implications

Lens progenitor cells have been derived from human ESCs as well as induced pluripotent stem cells (iPSCs) [48]. Lens stem cells are presumed to have a role in maintaining the lens transparency and might be important in cataractogenesis or other lens abnormalities

Retina

The retina represents the connecting link between visual input and image processing in the brain. Retinal diseases mostly result in irreversible damage to the visual pathway. Several studies in animal models have achieved some amount of success using transplantation of photoreceptors, endothelial cells and retinal pigment epithelium (RPE) [17, 48].

Therapeutic implications

Most therapeutic application studies have been conducted on murine retinal disease models. Diseases in the inner retina include retinopathy (ischemic conditions) and optic neuropathy, which cause damage in the retinal ganglion cells and amacrine cells [49]. Transplantation of bone marrow-derived mesenchymal stem cells into the vitreous of a retinal ischemia mouse model demonstrated ganglion cell neuroprotection [50]. Cell transplantation in a retinal degeneration model has shown promising visual outcomes but the extent of the curative effect remained unclear [51–55]. The injected stem cells integrated into the retinal and subretinal microenvironment modulated differentiation of different cell types [51, 52]. These transplanted cells integrate in a temporal-dependent manner that occurs only during rod genesis.

Clinical trials using fetal retinal cells have been conducted in patients with retinitis pigmentosa and age-related macular degeneration. In recent work, the entire retina has been replaced with differentiated stem cells rather than just single cells [53]. The three-dimensional neural retina was grown in culture from mouse ESCs [54]. Bilayer cups developed through morphogenesis of ESCs cultured with extracellular matrix. Most of the effects of transplantation seem to be based on the trophic factors used rather than a cell integration effect. Further studies and better sources of stem cells need to be investigated [55].

Photoreceptors and retinal-pigmented epithelium

The current stem cell-based therapies for retinal diseases focus on supplementing or replacing photoreceptors and RPE in the affected retina.

Therapeutic implications

Recently, safety and efficacy results were obtained from a clinical trial of subretinal transplantation of RPE cells derived from human ESCs. Several groups have shown the capacity of human ESCs to differentiate into RPE with variable success rates [56]. Recently, the differentiation efficacy was increased 30-fold by adding vitamin B3 and activin A protein [51]. Murine disease models such as Leber's congenital amaurosis rat have been used to study transplantation with differentiated retinal precursors; no teratoma formation was observed but the curative outcome needs to be followed up [57].

Transplantation of stem cell-, stem cell precursor- and iPSC-derived photoreceptors has resulted in functional recovery in animal models of retinal degeneration. Studies by several groups have demonstrated integration of photoreceptor precursors derived from postnatal retinas into degenerated mouse retina [52, 53, 58, 59]. Tucker and colleagues [60] demonstrated that adult fibroblast-derived iPSCs differentiated into retinal precursor cells expressing retinal as well as photoreceptor markers (Pax6, CRX, recoverin and rhodopsin). Moreover, research has progressed from differentiating ESCs into photoreceptor lineages to determining the type of cell and day of culture required for successful transplantation [61–63]. Investigating methods to improve and support transplantation, Tucker and colleagues [64] demonstrated that a xeno-free substrate and extracellular matrix-coated dishes resulted in similar differentiation of iPSCs to retinal cells.

Cultured RPE cells were transplanted into a rat model of age-related macular degeneration generated by defective photoreceptor phagocytosis [65]. The results revealed clearing of photoreceptor debris and regaining of visual function. Clinical trials on replacement of RPE in age-related macular degeneration resulted in transient vision recovery, with an autologous source providing better results [66].

Advanced Cell Technology Inc. (Marlborough, MA, USA) is currently conducting a phase I/II clinical trial on treating macular dystrophy using human ESC-derived RPE cells [67, 68]. Before RPE transplantation can be used in humans, further studies are necessary to determine how these cells can be integrated effectively into the retina without resulting in malignancy and immunogenicity [69].

Choroid

The choroid is derived from mesoderm and neuroectoderm. Choroidal stem cells obtained from murine studies reveal mesenchymal stem cell properties, expressing markers such as Sca-1, CD90.2, CD44, CD105, CD73, ABCG2, Six2, Notch1 and Pax6. We are still far from understanding their proliferative and differentiation potential [70].

Therapeutic implications

Choroidal and scleral cells have been differentiated into retinal lineage cells under laboratory conditions [71]. Further studies are needed to understand the biology as well as other attributes, such as differentiation and proliferative aspects, of these cells before they can be used clinically.

Sclera

The sclera is continuous with the cornea and is composed of fibrous material with viscoelastic properties. It is responsible for maintaining ocular pressure. Scleral stem cells have a mesenchymal origin and express ABCG2, Six2, Pax6 and Notch1 [70].

Therapeutic implications

Diseases that cause distension of the sclera, such as myopia, might be repaired using scleral stem cells. These might provide a source for bioengineering sclera for cell-based therapy [70], but more studies are needed to determine the role of scleral stem cells and treatment implications.

Orbit

The orbit provides a scaffold for the eye. It is a bony cavity that contains the eye, optic nerve, extra-ocular muscles, nerves, fat and lacrimal gland. In recent years, adipose tissue stem cells have been studied extensively, and some research has been done on orbital adipose stem cells. Diseases such as thyroid eye disease and aggressive malignant tumors lead to neurological consequences and blindness.

Identification and isolation

It has recently been observed that orbital fat cells contain stem cells. These are derived from neural crest cells, which are mesodermal in origin [72]. Interestingly, recent studies have isolated and characterized the orbital fat-derived stem cells. They have demonstrated that orbital adipocytes are similar to bone marrow-derived mesenchymal stem cells, sharing nearly 260 surface markers with them [73, 74]. Orbit fat-derived stem cells have the potential to differentiate into osteoblasts, chondrocytes and adipocytes, and further culturing with corneal epithelial cells changed their morphology to polygonal epithelial-like cells. This was confirmed by these cells expressing the epithelial cell marker zonal-occludin-1 and differentiation markers such as CK3 and CK19 [74].

Therapeutic implications

Destruction of corneal epithelial cells results in loss of vision. Stem cells isolated and expanded from the limbal area of the ocular surface are able to repair the corneal epithelium. However, obtaining healthy limbal stem cells and immune tolerance are still issues. Recently, Lin and colleagues [75] used orbital fat-derived stem cells to promote corneal tissue regeneration through a non-surgical route. Topical administration of fat-derived stem cells (mouse model) resulted in inhibition of inflammation and corneal re-epithelization. Therefore, orbit adipocytes are also potential candidates for cell therapy and tissue engineering of corneal epithelium. Further research into these progenitor cells may provide insight into pathological processes in orbit and other ocular damage.

Conclusion

We provide comprehensive detail on the localization of ocular stem cells and explain the therapeutic potential of each. Ocular diseases can be classified into vascular defects, anatomical defects and neurodegenerative defects. In order to address these defects, regenerative medicine using cell replacement strategies could be highly beneficial and effective. Identification of the proper sources of stem cells is the first step towards this, followed by their isolation and characterization. Ophthalmology is the only branch of medical science that has so far gained from the field of regenerative medicine. Limbal stem cell transplantation is the only other cell-based transplantation procedure, other than bone marrow transfusion, that has been approved for patient care.

Tables 1 and 2 provide summaries of the present and future prospects of stem cells for ocular therapy. Figure 1 depicts the locations of stem cells and their clinical application status. Figures 2 and 3 highlight stem cell sources used in ocular cell therapies for specific diseases. In order to harness the potential of stem cell-based therapy to provide and restore sight in blind patients, the safety of the cells needs to be studied in detail. For the successful utilization of stem cells for therapeutic purposes, small molecules can be incorporated with or conjugated to them before transplantation to promote specific differentiation pathways [76]. These cells serve to replace damaged cells and produce cytokines, growth factors, and other trophic molecules [77]. Fundamental studies are needed to unravel the roles of the Ivy league signaling pathways such as the Notch, WNT, Jak-Stat, tyrosine kinase, and Sonic hedgehog pathways. Also, alternative sources of stem cells need to be explored for their ability to integrate into the visual network. Basic researchers and ophthalmologists worldwide share optimism that stem cell therapy will in the future provide a means to restore vision.

Table 1 Ocular stem cells: locations, functions, markers and therapeutic development stage
Table 2 Future prospects for stem cell use for ocular cell therapy
Figure 1
figure 1

Schematic representation of sources and applications of ocular stem cells.

Figure 2
figure 2

Status of ocular and non-ocular stem cell transplantation for anterior surface disorders of the eye. MSC, mesenchymal stem cell.

Figure 3
figure 3

Status of ocular and non-ocular stem cell transplantation for posterior chamber disorders of the eye. MSC, mesenchymal stem cell.

Abbreviations

ABCG2:

ATP binding cassette sub family G member 2

CK:

Cytokeratin

ESC:

Embryonic stem cell

iPSC:

Induced pluripotent stem cell

LESC:

Limbal epithelial stem cell

RPE:

Retinal pigment epithelium

TM:

Trabecular meshwork.

References

  1. Blazejewska EA, Schlotzer-Schrehardt U, Zenkel M, Bachmann B, Chankiewitz E, Jacobi C, Kruse FE: Corneal limbal microenvironment can induce transdifferentiation of hair follicle stem cells into corneal epithelial-like cells. Stem Cells. 2009, 27: 642-652. 10.1634/stemcells.2008-0721.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  2. Ahmad I, Das AV, James J, Bhattacharya S, Zhao X: Neural stem cells in the mammalian eye: types and regulation. Semin Cell Dev Biol. 2004, 15: 53-62. 10.1016/j.semcdb.2003.09.003.

    Article  CAS  PubMed  Google Scholar 

  3. Abstracts of the 30th meeting of the Association for Eye Research including the annual meeting of the European Club for Ocular Fine Structure. Montpellier, France, 4–7 October 1989. Doc Ophthalmol. 1990, 76: 105-218.

  4. O'Sullivan F, Clynes M: Limbal stem cells, a review of their identification and culture for clinical use. Cytotechnology. 2007, 53: 101-106. 10.1007/s10616-007-9063-6.

    Article  PubMed Central  PubMed  Google Scholar 

  5. Daniels JT, Notara M, Shortt AJ, Secker G, Harris A, Tuft SJ: Limbal epithelial stem cell therapy. Expert Opin Biol Ther. 2007, 7: 1-3. 10.1517/14712598.7.1.1.

    Article  CAS  PubMed  Google Scholar 

  6. Cotsarelis G, Cheng SZ, Dong G, Sun TT, Lavker RM: Existence of slow-cycling limbal epithelial basal cells that can be preferentially stimulated to proliferate: implications on epithelial stem cells. Cell. 1989, 57: 201-209. 10.1016/0092-8674(89)90958-6.

    Article  CAS  PubMed  Google Scholar 

  7. Arpitha P, Prajna NV, Srinivasan M, Muthukkaruppan V: High expression of p63 combined with a large N/C ratio defines a subset of human limbal epithelial cells: implications on epithelial stem cells. Invest Ophthalmol Vis Sci. 2005, 46: 3631-3636. 10.1167/iovs.05-0343.

    Article  PubMed  Google Scholar 

  8. Pellegrini G, Rama P, Matuska S, Lambiase A, Bonini S, Pocobelli A, Colabelli RG, Spadea L, Fasciani R, Balestrazzi E, Vinciguerra P, Rosetta P, Tortori A, Nardi M, Gabbriellini G, Traverso CE, Macaluso C, Losi L, Percesepe A, Venturi B, Corradini F, Panaras A, Di Rocco A, Guatelli P, De Luca M: Biological parameters determining the clinical outcome of autologous cultures of limbal stem cells. Regen Med. 2013, 8: 553-567. 10.2217/rme.13.43.

    Article  CAS  PubMed  Google Scholar 

  9. Mariappan I, Maddileti S, Savy S, Tiwari S, Gaddipati S, Fatima A, Sangwan VS, Balasubramanian D, Vemuganti GK: In vitro culture and expansion of human limbal epithelial cells. Nat Protoc. 2010, 5: 1470-1479. 10.1038/nprot.2010.115.

    Article  CAS  PubMed  Google Scholar 

  10. Osei-Bempong C, Figueiredo FC, Lako M: The limbal epithelium of the eye - a review of limbal stem cell biology, disease and treatment. Bioessays. 2013, 35: 211-219. 10.1002/bies.201200086.

    Article  CAS  PubMed  Google Scholar 

  11. Chen Z, de Paiva CS, Luo L, Kretzer FL, Pflugfelder SC, Li DQ: Characterization of putative stem cell phenotype in human limbal epithelia. Stem Cells. 2004, 22: 355-366. 10.1634/stemcells.22-3-355.

    Article  PubMed Central  PubMed  Google Scholar 

  12. Dua HS, Joseph A, Shanmuganathan VA, Jones RE: Stem cell differentiation and the effects of deficiency. Eye (Lond). 2003, 17: 877-885. 10.1038/sj.eye.6700573.

    Article  CAS  Google Scholar 

  13. Amano S, Yamagami S, Mimura T, Uchida S, Yokoo S: Corneal stromal and endothelial cell precursors. Cornea. 2006, 25: S73-S77. 10.1097/01.ico.0000247218.10672.7e.

    Article  PubMed  Google Scholar 

  14. Yoshida S, Shimmura S, Nagoshi N, Fukuda K, Matsuzaki Y, Okano H, Tsubota K: Isolation of multipotent neural crest-derived stem cells from the adult mouse cornea. Stem Cells. 2006, 24: 2714-2722. 10.1634/stemcells.2006-0156.

    Article  CAS  PubMed  Google Scholar 

  15. Pinnamaneni N, Funderburgh JL: Concise review: Stem cells in the corneal stroma. Stem Cells. 2012, 30: 1059-1063. 10.1002/stem.1100.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  16. Du Y, Funderburgh ML, Mann MM, SundarRaj N, Funderburgh JL: Multipotent stem cells in human corneal stroma. Stem Cells. 2005, 23: 1266-1275. 10.1634/stemcells.2004-0256.

    Article  PubMed Central  PubMed  Google Scholar 

  17. Pellegrini G, De Luca M, Arsenijevic Y: Towards therapeutic application of ocular stem cells. Semin Cell Dev Biol. 2007, 18: 805-818. 10.1016/j.semcdb.2007.09.011.

    Article  CAS  PubMed  Google Scholar 

  18. Pellegrini G, Golisano O, Paterna P, Lambiase A, Bonini S, Rama P, De Luca M: Location and clonal analysis of stem cells and their differentiated progeny in the human ocular surface. J Cell Biol. 1999, 145: 769-782. 10.1083/jcb.145.4.769.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. Priya CG, Arpitha P, Vaishali S, Prajna NV, Usha K, Sheetal K, Muthukkaruppan V: Adult human buccal epithelial stem cells: identification, ex-vivo expansion, and transplantation for corneal surface reconstruction. Eye (Lond). 2011, 25: 1641-1649. 10.1038/eye.2011.230.

    Article  CAS  Google Scholar 

  20. Sangwan VS, Matalia HP, Vemuganti GK, Fatima A, Ifthekar G, Singh S, Nutheti R, Rao GN: Clinical outcome of autologous cultivated limbal epithelium transplantation. Indian J Ophthalmol. 2006, 54: 29-34. 10.4103/0301-4738.21611.

    Article  PubMed  Google Scholar 

  21. Mimura T, Tabata Y, Amano S: Transplantation of corneal stroma reconstructed with gelatin and multipotent precursor cells from corneal stroma. Tissue Engineering for Tissue and Organ Regeneration. Edited by: Eberli D. 2010, : Rijeka, InTech, 347-362.

    Google Scholar 

  22. Du Y, Carlson EC, Funderburgh ML, Birk DE, Pearlman E, Guo N, Kao WW, Funderburgh JL: Stem cell therapy restores transparency to defective murine corneas. Stem Cells. 2009, 27: 1635-1642. 10.1002/stem.91.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  23. Qi H, Zheng X, Yuan X, Pflugfelder SC, Li DQ: Potential localization of putative stem/progenitor cells in human bulbar conjunctival epithelium. J Cell Physiol. 2010, 225: 180-185. 10.1002/jcp.22215.

    Article  CAS  PubMed  Google Scholar 

  24. Rosellini A, Papini S, Giannarini C, Nardi M, Revoltella RP: Human conjunctival epithelial precursor cells and their progeny in 3D organotypic culture. Int J Dev Biol. 2007, 51: 739-743. 10.1387/ijdb.062198ar.

    Article  CAS  PubMed  Google Scholar 

  25. Tanioka H, Kawasaki S, Yamasaki K, Ang LP, Koizumi N, Nakamura T, Yokoi N, Komuro A, Inatomi T, Kinoshita S: Establishment of a cultivated human conjunctival epithelium as an alternative tissue source for autologous corneal epithelial transplantation. Invest Ophthalmol Vis Sci. 2006, 47: 3820-3827. 10.1167/iovs.06-0293.

    Article  PubMed  Google Scholar 

  26. Ricardo JR, Cristovam PC, Filho PA, Farias CC, de Araujo AL, Loureiro RR, Covre JL, de Barros JN, Barreiro TP, dos Santos MS, Gomes JA: Transplantation of conjunctival epithelial cells cultivated ex vivo in patients with total limbal stem cell deficiency. Cornea. 2013, 32: 221-228. 10.1097/ICO.0b013e31825034be.

    Article  PubMed  Google Scholar 

  27. Ang LP, Cheng ZY, Beuerman RW, Teoh SH, Zhu X, Tan DT: The development of a serum-free derived bioengineered conjunctival epithelial equivalent using an ultrathin poly(epsilon-caprolactone) membrane substrate. Invest Ophthalmol Vis Sci. 2006, 47: 105-112. 10.1167/iovs.05-0512.

    Article  PubMed  Google Scholar 

  28. Grierson I, Pfeiffer N, Cracknell KP, Appleton P: Histology and fine structure of the iris and outflow system following latanoprost therapy. Surv Ophthalmol. 2002, 47: S176-S184.

    Article  PubMed  Google Scholar 

  29. Arnhold S, Semkova I, Andressen C, Lenartz D, Meissner G, Sturm V, Kochanek S, Addicks K, Schraermeyer U: Iris pigment epithelial cells: a possible cell source for the future treatment of neurodegenerative diseases. Exp Neurol. 2004, 187: 410-417. 10.1016/j.expneurol.2004.02.015.

    Article  CAS  PubMed  Google Scholar 

  30. Seko Y, Azuma N, Kaneda M, Nakatani K, Miyagawa Y, Noshiro Y, Kurokawa R, Okano H, Umezawa A: Derivation of human differential photoreceptor-like cells from the iris by defined combinations of CRX, RX and NEUROD. PLoS One. 2012, 7: e35611-10.1371/journal.pone.0035611.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  31. Napier HR, Kidson SH: Molecular events in early development of the ciliary body: a question of folding. Exp Eye Res. 2007, 84: 615-625. 10.1016/j.exer.2006.07.012.

    Article  CAS  PubMed  Google Scholar 

  32. Xu H, Sta Iglesia DD, Kielczewski JL, Valenta DF, Pease ME, Zack DJ, Quigley HA: Characteristics of progenitor cells derived from adult ciliary body in mouse, rat, and human eyes. Invest Ophthalmol Vis Sci. 2007, 48: 1674-1682. 10.1167/iovs.06-1034.

    Article  PubMed  Google Scholar 

  33. Gualdoni S, Baron M, Lakowski J, Decembrini S, Smith AJ, Pearson RA, Ali RR, Sowden JC: Adult ciliary epithelial cells, previously identified as retinal stem cells with potential for retinal repair, fail to differentiate into new rod photoreceptors. Stem Cells. 2010, 28: 1048-1059. 10.1002/stem.423.

    Article  CAS  PubMed  Google Scholar 

  34. Yanagi Y, Inoue Y, Kawase Y, Uchida S, Tamaki Y, Araie M, Okochi H: Properties of growth and molecular profiles of rat progenitor cells from ciliary epithelium. Exp Eye Res. 2006, 82: 471-478. 10.1016/j.exer.2005.08.005.

    Article  CAS  PubMed  Google Scholar 

  35. Cicero SA, Johnson D, Reyntjens S, Frase S, Connell S, Chow LM, Baker SJ, Sorrentino BP, Dyer MA: Cells previously identified as retinal stem cells are pigmented ciliary epithelial cells. Proc Natl Acad Sci U S A. 2009, 106: 6685-6690. 10.1073/pnas.0901596106.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  36. Ballios BG, Clarke L, Coles BL, Shoichet MS, Van Der Kooy D: The adult retinal stem cell is a rare cell in the ciliary epithelium whose progeny can differentiate into photoreceptors. Biol Open. 2012, 1: 237-246. 10.1242/bio.2012027.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  37. Inoue T, Coles BL, Dorval K, Bremner R, Bessho Y, Kageyama R, Hino S, Matsuoka M, Craft CM, McInnes RR, Tremblay F, Prusky GT, van der Kooy D: Maximizing functional photoreceptor differentiation from adult human retinal stem cells. Stem Cells. 2010, 28: 489-500.

    PubMed Central  CAS  PubMed  Google Scholar 

  38. Du Y, Roh DS, Mann MM, Funderburgh ML, Funderburgh JL, Schuman JS: Multipotent stem cells from trabecular meshwork become phagocytic TM cells. Invest Ophthalmol Vis Sci. 2012, 53: 1566-1575. 10.1167/iovs.11-9134.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  39. Gonzalez P, Caballero M, Liton PB, Stamer WD, Epstein DL: Expression analysis of the matrix GLA protein and VE-cadherin gene promoters in the outflow pathway. Invest Ophthalmol Vis Sci. 2004, 45: 1389-1395. 10.1167/iovs.03-0537.

    Article  PubMed  Google Scholar 

  40. Liton PB, Liu X, Stamer WD, Challa P, Epstein DL, Gonzalez P: Specific targeting of gene expression to a subset of human trabecular meshwork cells using the chitinase 3-like 1 promoter. Invest Ophthalmol Vis Sci. 2005, 46: 183-190. 10.1167/iovs.04-0330.

    Article  PubMed Central  PubMed  Google Scholar 

  41. Stamer WD, Seftor RE, Snyder RW, Regan JW: Cultured human trabecular meshwork cells express aquaporin-1 water channels. Curr Eye Res. 1995, 14: 1095-1100. 10.3109/02713689508995815.

    Article  CAS  PubMed  Google Scholar 

  42. Tay CY, Sathiyanathan P, Chu SW, Stanton LW, Wong TT: Identification and characterization of mesenchymal stem cells derived from the trabecular meshwork of the human eye. Stem Cells Dev. 2012, 21: 1381-1390. 10.1089/scd.2011.0655.

    Article  CAS  PubMed  Google Scholar 

  43. Acott TS, Samples JR, Bradley JM, Bacon DR, Bylsma SS, Van Buskirk EM: Trabecular repopulation by anterior trabecular meshwork cells after laser trabeculoplasty. Am J Ophthalmol. 1989, 107: 1-6.

    Article  CAS  PubMed  Google Scholar 

  44. Du Y, Yun H, Yang E, Schuman JS: Stem cells from trabecular meshwork home to TM tissue in vivo. Invest Ophthalmol Vis Sci. 2013, 54: 1450-1459. 10.1167/iovs.12-11056.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  45. Yu WY, Sheridan C, Grierson I, Mason S, Kearns V, Lo AC, Wong D: Progenitors for the corneal endothelium and trabecular meshwork: a potential source for personalized stem cell therapy in corneal endothelial diseases and glaucoma. J Biomed Biotechnol. 2011, 2011: 412743-

    Article  PubMed Central  PubMed  Google Scholar 

  46. Remington SG, Meyer RA: Lens stem cells may reside outside the lens capsule: an hypothesis. Theor Biol Med Model. 2007, 4: 22-10.1186/1742-4682-4-22.

    Article  PubMed Central  PubMed  Google Scholar 

  47. Tsonis PA, Del Rio-Tsonis K: Lens and retina regeneration: transdifferentiation, stem cells and clinical applications. Exp Eye Res. 2004, 78: 161-172. 10.1016/j.exer.2003.10.022.

    Article  CAS  PubMed  Google Scholar 

  48. Qiu X, Yang J, Liu T, Jiang Y, Le Q, Lu Y: Efficient generation of lens progenitor cells from cataract patient-specific induced pluripotent stem cells. PLoS One. 2012, 7: e32612-10.1371/journal.pone.0032612.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Laouri M, Chen E, Looman M, Gallagher M: The burden of disease of retinal vein occlusion: review of the literature. Eye (Lond). 2011, 25: 981-988. 10.1038/eye.2011.92.

    Article  CAS  Google Scholar 

  50. Li N, Li XR, Yuan JQ: Effects of bone-marrow mesenchymal stem cells transplanted into vitreous cavity of rat injured by ischemia/reperfusion. Graefes Arch Clin Exp Ophthalmol. 2009, 247: 503-514. 10.1007/s00417-008-1009-y.

    Article  PubMed  Google Scholar 

  51. Banin E, Obolensky A, Idelson M, Hemo I, Reinhardtz E, Pikarsky E, Ben-Hur T, Reubinoff B: Retinal incorporation and differentiation of neural precursors derived from human embryonic stem cells. Stem Cells. 2006, 24: 246-257. 10.1634/stemcells.2005-0009.

    Article  PubMed  Google Scholar 

  52. MacLaren RE, Pearson RA, MacNeil A, Douglas RH, Salt TE, Akimoto M, Swaroop A, Sowden JC, Ali RR: Retinal repair by transplantation of photoreceptor precursors. Nature. 2006, 444: 203-207. 10.1038/nature05161.

    Article  CAS  PubMed  Google Scholar 

  53. Ali RR, Sowden JC: Regenerative medicine: DIY eye. Nature. 2011, 472: 42-43. 10.1038/472042a.

    Article  CAS  PubMed  Google Scholar 

  54. Eiraku M, Takata N, Ishibashi H, Kawada M, Sakakura E, Okuda S, Sekiguchi K, Adachi T, Sasai Y: Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature. 2011, 472: 51-56. 10.1038/nature09941.

    Article  CAS  PubMed  Google Scholar 

  55. Radtke ND, Aramant RB, Seiler MJ, Petry HM, Pidwell D: Vision change after sheet transplant of fetal retina with retinal pigment epithelium to a patient with retinitis pigmentosa. Arch Ophthalmol. 2004, 122: 1159-1165. 10.1001/archopht.122.8.1159.

    Article  PubMed  Google Scholar 

  56. Tibbetts MD, Samuel MA, Chang TS, Ho AC: Stem cell therapy for retinal disease. Curr Opin Ophthalmol. 2012, 23: 226-234. 10.1097/ICU.0b013e328352407d.

    Article  PubMed  Google Scholar 

  57. Lamba DA, McUsic A, Hirata RK, Wang PR, Russell D, Reh TA: Generation, purification and transplantation of photoreceptors derived from human induced pluripotent stem cells. PLoS One. 2010, 5: e8763-10.1371/journal.pone.0008763.

    Article  PubMed Central  PubMed  Google Scholar 

  58. Barber AC, Hippert C, Duran Y, West EL, Bainbridge JW, Warre-Cornish K, Luhmann UF, Lakowski J, Sowden JC, Ali RR, Pearson RA: Repair of the degenerate retina by photoreceptor transplantation. Proc Natl Acad Sci U S A. 2013, 110: 354-359. 10.1073/pnas.1212677110.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  59. Singh MS, Charbel Issa P, Butler R, Martin C, Lipinski DM, Sekaran S, Barnard AR, MacLaren RE: Reversal of end-stage retinal degeneration and restoration of visual function by photoreceptor transplantation. Proc Natl Acad Sci U S A. 2013, 110: 1101-1106. 10.1073/pnas.1119416110.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  60. Tucker BA, Park IH, Qi SD, Klassen HJ, Jiang C, Yao J, Redenti S, Daley GQ, Young MJ: Transplantation of adult mouse iPS cell-derived photoreceptor precursors restores retinal structure and function in degenerative mice. PLoS One. 2011, 6: e18992-10.1371/journal.pone.0018992.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  61. Osakada F, Ikeda H, Mandai M, Wataya T, Watanabe K, Yoshimura N, Akaike A, Sasai Y, Takahashi M: Toward the generation of rod and cone photoreceptors from mouse, monkey and human embryonic stem cells. Nat Biotechnol. 2008, 26: 215-224. 10.1038/nbt1384.

    Article  CAS  PubMed  Google Scholar 

  62. West EL, Gonzalez-Cordero A, Hippert C, Osakada F, Martinez-Barbera JP, Pearson RA, Sowden JC, Takahashi M, Ali RR: Defining the integration capacity of embryonic stem cell-derived photoreceptor precursors. Stem Cells. 2012, 30: 1424-1435. 10.1002/stem.1123.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  63. Gonzalez-Cordero A, West EL, Pearson RA, Duran Y, Carvalho LS, Chu CJ, Naeem A, Blackford SJ, Georgiadis A, Lakowski J, Hubank M, Smith AJ, Bainbridge JW, Sowden JC, Ali RR: Photoreceptor precursors derived from three-dimensional embryonic stem cell cultures integrate and mature within adult degenerate retina. Nat Biotechnol. 2013, 31: 741-747. 10.1038/nbt.2643.

    Article  CAS  PubMed  Google Scholar 

  64. Tucker BA, Anfinson KR, Mullins RF, Stone EM, Young MJ: Use of a synthetic xeno-free culture substrate for induced pluripotent stem cell induction and retinal differentiation. Stem Cells Transl Med. 2013, 2: 16-24. 10.5966/sctm.2012-0040.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  65. Lund RD, Kwan AS, Keegan DJ, Sauve Y, Coffey PJ, Lawrence JM: Cell transplantation as a treatment for retinal disease. Prog Retin Eye Res. 2001, 20: 415-449. 10.1016/S1350-9462(01)00003-9.

    Article  CAS  PubMed  Google Scholar 

  66. MacLaren RE, Bird AC, Sathia PJ, Aylward GW: Long-term results of submacular surgery combined with macular translocation of the retinal pigment epithelium in neovascular age-related macular degeneration. Ophthalmology. 2005, 112: 2081-2087. 10.1016/j.ophtha.2005.06.029.

    Article  PubMed  Google Scholar 

  67. Safety and Tolerability of Sub-retinal Transplantation of Human Embryonic Stem Cell Derived Retinal Pigmented Epithelial (hESC-RPE) Cells in Patients With Stargardt's Macular Dystrophy (SMD). A Phase I/II, Open-Label, Multi-Center interventional trial. http://clinicaltrial.gov/ct2/show/NCT01469832]

  68. Sub-retinal Transplantation of hESC Derived RPE (MA09-hRPE) Cells in Patients With Stargardt's Macular Dystrophy. A Phase I/II, Open-Label, Multi-Center interventional trial. http://clinicaltrial.gov/ct2/show/NCT01345006]

  69. Lakowski J, Han YT, Pearson RA, Gonzalez-Cordero A, West EL, Gualdoni S, Barber AC, Hubank M, Ali RR, Sowden JC: Effective transplantation of photoreceptor precursor cells selected via cell surface antigen expression. Stem Cells. 2011, 29: 1391-1404.

    PubMed Central  CAS  PubMed  Google Scholar 

  70. Tsai CL, Wu PC, Fini ME, Shi S: Identification of multipotent stem/progenitor cells in murine sclera. Invest Ophthalmol Vis Sci. 2011, 52: 5481-5487. 10.1167/iovs.11-7676.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  71. Arsenijevic Y, Taverney N, Kostic C, Tekaya M, Riva F, Zografos L, Schorderet D, Munier F: Non-neural regions of the adult human eye: a potential source of neurons?. Invest Ophthalmol Vis Sci. 2003, 44: 799-807. 10.1167/iovs.02-0267.

    Article  PubMed  Google Scholar 

  72. Johnston MC, Noden DM, Hazelton RD, Coulombre JL, Coulombre AJ: Origins of avian ocular and periocular tissues. Exp Eye Res. 1979, 29: 27-43. 10.1016/0014-4835(79)90164-7.

    Article  CAS  PubMed  Google Scholar 

  73. Ho JH, Ma WH, Tseng TC, Chen YF, Chen MH, Lee OK: Isolation and characterization of multi-potent stem cells from human orbital fat tissues. Tissue Eng Part A. 2011, 17: 255-266. 10.1089/ten.tea.2010.0106.

    Article  CAS  PubMed  Google Scholar 

  74. Chien MH, Bien MY, Ku CC, Chang YC, Pao HY, Yang YL, Hsiao M, Chen CL, Ho JH: Systemic human orbital fat-derived stem/stromal cell transplantation ameliorates acute inflammation in lipopolysaccharide-induced acute lung injury. Crit Care Med. 2012, 40: 1245-1253. 10.1097/CCM.0b013e31823bc89a.

    Article  CAS  PubMed  Google Scholar 

  75. Lin KJ, Loi MX, Lien GS, Cheng CF, Pao HY, Chang YC, Ji AT, Ho JH: Topical administration of orbital fat-derived stem cells promotes corneal tissue regeneration. Stem Cell Res Ther. 2013, 4: 72-10.1186/scrt223.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  76. Romano AC, Espana EM, Yoo SH, Budak MT, Wolosin JM, Tseng SC: Different cell sizes in human limbal and central corneal basal epithelia measured by confocal microscopy and flow cytometry. Invest Ophthalmol Vis Sci. 2003, 44: 5125-5129. 10.1167/iovs.03-0628.

    Article  PubMed  Google Scholar 

  77. Sengupta N, Caballero S, Sullivan SM, Chang LJ, Afzal A, Li Calzi S, Kielczewski JL, Prabarakan S, Ellis EA, Moldovan L, Moldovan NI, Boulton ME, Grant MB, Scott EW, Harris JR: Regulation of adult hematopoietic stem cells fate for enhanced tissue-specific repair. Mol Ther. 2009, 17: 1594-1604. 10.1038/mt.2009.145.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  78. Lappas A, Foerster AM, Weinberger AW, Coburger S, Schrage NF, Kirchhof B: Translocation of iris pigment epithelium in patients with exudative age-related macular degeneration: long-term results. Graefes Arch Clin Exp Ophthalmol. 2004, 242: 638-647. 10.1007/s00417-003-0764-z.

    Article  PubMed  Google Scholar 

  79. Aisenbrey S, Lafaut BA, Szurman P, Hilgers RD, Esser P, Walter P, Bartz-Schmidt KU, Thumann G: Iris pigment epithelial translocation in the treatment of exudative macular degeneration: a 3-year follow-up. Arch Ophthalmol. 2006, 124: 183-188. 10.1001/archopht.124.2.183.

    Article  PubMed  Google Scholar 

  80. Lee JY, Lee KH, Shin HM, Chung KH, Kim GI, Lew H: Orbital volume augmentation after injection of human orbital adipose-derived stem cells in rabbits. Invest Ophthalmol Vis Sci. 2013, 54: 2410-2416. 10.1167/iovs.12-11210.

    Article  PubMed  Google Scholar 

  81. Hynes SR, Lavik EB: A tissue-engineered approach towards retinal repair: scaffolds for cell transplantation to the subretinal space. Graefes Arch Clin Exp Ophthalmol. 2010, 248: 763-778. 10.1007/s00417-009-1263-7.

    Article  PubMed  Google Scholar 

  82. Sodha S, Wall K, Redenti S, Klassen H, Young MJ, Tao SL: Microfabrication of a three-dimensional polycaprolactone thin-film scaffold for retinal progenitor cell encapsulation. J Biomater Sci Polym Ed. 2011, 22: 443-456. 10.1163/092050610X487738.

    Article  CAS  PubMed  Google Scholar 

  83. McUsic AC, Lamba DA, Reh TA: Guiding the morphogenesis of dissociated newborn mouse retinal cells and hES cell-derived retinal cells by soft lithography-patterned microchannel PLGA scaffolds. Biomaterials. 2012, 33: 1396-1405. 10.1016/j.biomaterials.2011.10.083.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  84. Thieltges F, Stanzel BV, Liu Z, Holz FG: A nanofibrillar surface promotes superior growth characteristics in cultured human retinal pigment epithelium. Ophthalmic Res. 2011, 46: 133-140. 10.1159/000324045.

    Article  CAS  PubMed  Google Scholar 

  85. Meyer JS, Howden SE, Wallace KA, Verhoeven AD, Wright LS, Capowski EE, Pinilla I, Martin JM, Tian S, Stewart R, Pattnaik B, Thomson JA, Gamm DM: Optic vesicle-like structures derived from human pluripotent stem cells facilitate a customized approach to retinal disease treatment. Stem Cells. 2011, 29: 1206-1218. 10.1002/stem.674.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The authors would like to convey their sincere gratitude to Dr K Bhujang Shetty, Dr Rohit Shetty and Dr Himanshu Matalia. The authors would like to thank the Narayana Nethralaya Foundation, India, Department of Science and Technology, Government of India and the Discovery Eye Foundation/National Keratoconus Foundation, USA, for providing all the necessary support for this review. KD is supported by the Department of Science and Technology for Senior Research Fellowship. We would also like to thank Neeraj Bhas for work on the figures.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Debashish Das.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ original submitted files for images

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dhamodaran, K., Subramani, M., Ponnalagu, M. et al. Ocular stem cells: a status update!. Stem Cell Res Ther 5, 56 (2014). https://doi.org/10.1186/scrt445

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/scrt445

Keywords