Next Article in Journal
Age-Related Macular Degeneration: Advances in Management and Diagnosis
Next Article in Special Issue
Using iPS Cells toward the Understanding of Parkinson’s Disease
Previous Article in Journal
Association of OCT-Derived Drusen Measurements with AMD-Associated Genotypic SNPs in the Amish Population
Previous Article in Special Issue
Myogenic Precursors from iPS Cells for Skeletal Muscle Cell Replacement Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Role of Induced Pluripotent Stem Cells (IPSCs) for Cell-Based Therapy of the Ocular Surface

by
Ricardo P. Casaroli-Marano
1,2,3,*,
Núria Nieto-Nicolau
2,
Eva M. Martínez-Conesa
3,
Michael Edel
4,5 and
Ana B.Álvarez-Palomo
4
1
Department of Surgery, School of Medicine and Hospital Clínic de Barcelona (IDIBAPS), University of Barcelona, Calle Sabino de Arana 1 (2nd floor), E-08028 Barcelona, Spain
2
CellTec-UB and the Clinic Foundation for Biomedical Research (FCRB), University of Barcelona, Avda. Diagonal 643, E-08028 Barcelona, Spain
3
Tissue Bank of BST (GenCat), Calle Dr Antoni Pujadas 42, SSMM Sant Joan de Déu, Edifici Pujadas, E-08830 Sant Boi de Llobregat, Spain
4
Pluripotency Group, Department of Physiology I, School of Medicine, University of Barcelona, Calle Casanovas 143, E-08036 Barcelona, Spain
5
Faculty of Medicine, Children's Hospital, University of Sydney, 2006 NSW, Australia
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2015, 4(2), 318-342; https://doi.org/10.3390/jcm4020318
Submission received: 23 November 2014 / Revised: 24 December 2014 / Accepted: 4 January 2015 / Published: 12 February 2015
(This article belongs to the Special Issue iPS Cells for Modelling and Treatment of Human Diseases)

Abstract

:
The integrity and normal function of the corneal epithelium are crucial for maintaining the cornea’s transparency and vision. The existence of a cell population with progenitor characteristics in the limbus maintains a dynamic of constant epithelial repair and renewal. Currently, cell-based therapies for bio replacement—cultured limbal epithelial transplantation (CLET) and cultured oral mucosal epithelial transplantation (COMET)—present very encouraging clinical results for treating limbal stem cell deficiency (LSCD) and restoring vision. Another emerging therapeutic approach consists of obtaining and implementing human progenitor cells of different origins in association with tissue engineering methods. The development of cell-based therapies using stem cells, such as human adult mesenchymal or induced pluripotent stem cells (IPSCs), represent a significant breakthrough in the treatment of certain eye diseases, offering a more rational, less invasive, and better physiological treatment option in regenerative medicine for the ocular surface. This review will focus on the main concepts of cell-based therapies for the ocular surface and the future use of IPSCs to treat LSCD.

1. Introduction

The ocular surface is mainly composed of the cornea and the conjunctiva with their epithelia. The cornea is the primary refractive element at the anterior surface of the eye that is responsible for approximately two-thirds of its total optical power. Basically, the cornea is composed of five well-defined layers (Figure 1). It consists of an outermost stratified, squamous and non-keratinized epithelial layer (corneal epithelium) limited posteriorly by Bowman’s layer. The underlying stroma, which accounts for about 90% of the middle thickness of the cornea, comprises aligned arrays of collagen fibrils interspersed with cellular components (keratocytes) and it is this highly organized arrangement of lamellae that is responsible for the cornea’s transparency. The stroma is separated from the endothelial layer (corneal endothelium) by Descemet’s membrane, which acts as a basement membrane for these endothelial cells. The corneal endothelium is a single cuboidal layer of metabolically active cells that are in direct contact with the aqueous humor in the anterior chamber. These cells help to maintain corneal transparency by actively pumping water out of the stroma [1]. The corneal epithelium has a key role in keeping the cornea transparent and free of blood vessels and, to this end, presents permanent repair phenomena essential for the conservation of the cornea’s physiology [1,2,3]. The homeostasis of the corneal epithelium is crucial to maintaining the structural integrity of the ocular surface, the transparency of the cornea and visual function.

1.1. Limbal Stem Cells

It has been observed that progenitor cells responsible for the continual renewal of the corneal epithelium are located in the basal layers of the sclerocorneal limbus. The human limbus—the circumferential anatomic area (approximately 1.5 mm wide) that separates the clear cornea from the opaque sclera, which is covered by conjunctiva—serves as the “reservoir” for the stem cells and also provides a barrier to the overgrowth of conjunctival epithelial cells and its blood vessels onto the cornea [1,2,3] (Figure 1). Due to their particularities, the limbal stem cells (LSCs) have a crucial role in maintaining the integrity and in the renewal events of corneal epithelium. Their main features are highlighted: it is their behavior as oligopotent progenitor cells, with high nuclear-cytoplasmic ratio a slow cell cycle, and a high proliferative potential that adds its great capacity for self-renewal by asymmetric division [3,4,5]. In the limbus, it is possible to identify several cell subpopulations of different progenies (typical progenitors and amplifying cells at different stages of differentiation), melanocytes, antigen-presenting and mesenchymal cells, vascular elements and nerve endings that form a specialized and unique environment called niche. This particular microenvironment is considered responsible for the proliferative and self-renewal cellular characteristics of the limbal region [2,3,6]. The LSC niche is an anatomically defined area that is thought to provide a variety of factors, such as physical protection, survival factors and cytokines and is deemed essential to the maintenance of the “stemness” of the stem cell population while preventing entry into differentiation [2,6]. Within the niche, LSCs maintenance and function are controlled in a particular environment by several elements, including extracellular matrix components, cell adhesion molecules, and growth and survival factors secreted by stromal fibroblasts, mesenchymal stem cells and blood capillaries [6]. To date, four limbal anatomic structures have been proposed as the corneal stem cell niche [2,6], Palisades of Vogt, limbal epithelial crypts [7], limbal crypts and focal stromal projections [8].
Figure 1. The corneal limbus is the circumferential anatomic area, approximately 1.5 mm wide, which separates the clear cornea from the opaque sclera (a); The limbal region represents the “reservoir” for LSCs in the ocular surface. In a cross-section of the human cornea stained with hematoxylin-eosin, (b) to (d), details of its main layers can be observed. The cornea is composed of a stratified non-keratinized squamous epithelial layer (epithelium), the stroma and an endothelial cuboidal layer (endothelium) (b); The corneal epithelium (48 to 55 µm thick) consists of the outermost layer, which presents five to seven stratified cell layers (c), limited posteriorly by Bowman’s layer (10 to 12 µm thick; c, asterisk). The stroma (480 to 510 µm thick; b), composed of compacted collagen lamellae and keratocytes (c and d), offers transparency and scaffolding to maintain the shape of the cornea in its middle portion. The stroma is separated from the endothelium (about 5 µm thick; d, large arrows) by Descemet’s membrane (8 to 10 µm thick; d, narrow arrows), which acts as a basement membrane for the corneal endothelial cells (d). Bar = 150 µm for b; Bar = 25 µm for c and d.
Figure 1. The corneal limbus is the circumferential anatomic area, approximately 1.5 mm wide, which separates the clear cornea from the opaque sclera (a); The limbal region represents the “reservoir” for LSCs in the ocular surface. In a cross-section of the human cornea stained with hematoxylin-eosin, (b) to (d), details of its main layers can be observed. The cornea is composed of a stratified non-keratinized squamous epithelial layer (epithelium), the stroma and an endothelial cuboidal layer (endothelium) (b); The corneal epithelium (48 to 55 µm thick) consists of the outermost layer, which presents five to seven stratified cell layers (c), limited posteriorly by Bowman’s layer (10 to 12 µm thick; c, asterisk). The stroma (480 to 510 µm thick; b), composed of compacted collagen lamellae and keratocytes (c and d), offers transparency and scaffolding to maintain the shape of the cornea in its middle portion. The stroma is separated from the endothelium (about 5 µm thick; d, large arrows) by Descemet’s membrane (8 to 10 µm thick; d, narrow arrows), which acts as a basement membrane for the corneal endothelial cells (d). Bar = 150 µm for b; Bar = 25 µm for c and d.
Jcm 04 00318 g001

1.2. Renewal of Corneal Epithelium

It has been shown that cell subpopulations with progenitor features, located in the deeper basal layers of the corneal epithelium, have the capacity to differentiate into post-mitotic cell populations located in the outermost epithelial layers. This continuous centripetal movement (the XYZ hypothesis)—from the peripheral deeper epithelial layers to the more central outermost layers—ensures constant renewal of the corneal epithelium and maintains its integrity [1,2,3,4,5,6,9]. The X component represents the anterior migration from cells of the basal epithelium of the limbal region, the Y component represents the centripetal migration of cells from the limbus, and the Z component represents the desquamation from the surface of corneal epithelium. However, this XYZ theory has recently been challenged by evidence in the mouse and other mammals suggesting that uninjured cells in the central cornea can generate holoclones with characteristics of stem cells, presenting regenerative epithelial capabilities, which may also be responsible for the maintenance of the corneal epithelium [10]. Also, in support of these controversial findings, the presence of central islands of normal corneal epithelial cells has been described in patients with apparently complete clinical absence of LSCs [11]. These interesting observations may have the following interpretation: the central basal epithelial cells of the surviving corneal epithelium present the capability to regenerate, or some LSCs remain and contribute to the maintenance of the central epithelium.

1.3. Limbal Stem Cell Multipotency

An in vitro study of the clonogenic capacity of epithelial cells located in the ocular limbal region revealed a progenitor cell system stratified into levels (cellular stages or “compartments”) [2,6,12]. Undifferentiated small cells presenting progenitor cell features with high self-renewal capacity are found in the first compartment but they lose these characteristics as they migrate through the following compartments. Lastly, the final level contains a cell population with terminal differentiation features associated with little or no self-renewal capability. The latter cells, once their epithelial differentiation events are completed, lose their ability to self-renew and are incorporated as corneal epithelial cells on the surface of the central cornea. In this regard, some studies [12,13] concluded that epithelial cells of the limbal region can form holoclones with higher clonogenic potential, in contrast to epithelial cells from the central cornea. In addition, epithelial cells isolated from basal layers in the limbal region exhibit a high proliferative potential in vitro during expansion or in response to corneal injury [14], and show an undifferentiated phenotype lacking the expression of differentiated corneal cell markers such as cytokeratins 3 and 12 [15]. They have also been shown to retain labeled precursors of DNA for an extended time, in contrast to more differentiated cells that quickly lose them due a higher division rate [16]. This lack of differentiation and slow cell cycling are characteristics of the quiescent state of stem cells.

2. Limbal Stem Cell Deficiency (LSCD)

The disappearance, reduction or functional impairment of LSCs may produce a clinical state (limbal stem cell deficiency, LSCD) that can give rise to significant changes in the ocular surface. These changes include the occurrence of persistent corneal defects, epithelial keratinization, conjunctivalization phenomena with the development of newly formed vessels in the corneal tissue, and scarring. All this compromises the corneal physiology, reducing transparency and decreasing vision [1,2,3,4,5]. The presence of a complete loss of the corneal-limbal epithelium leads to a reactive reepithelialization by conjunctival cells, which have a high proliferative capacity. This event is followed by neovascularization, chronic inflammation with scarring of corneal stroma, causing a pronounced decrease in vision and severe discomfort (Figure 2). Furthermore, the chronic inflammatory condition not only leads to the death of more LSCs but also leaves the surviving epithelial cells unable to function properly, explaining the worsening of clinical symptoms and features over time [17,18,19,20,21,22,23,24]. In patients with severe lacrimal dysfunction syndrome (dry eye) suffering from LSCD, the conjunctival epithelium that replaced the corneal epithelium (conjunctivalization) becomes partially or totally keratinized [18,21,22,24]. Several processes and diseases (Table 1) may lead to unilateral or bilateral LSCD, and depending on its extent, the disorder can be classified as either partial or total. Chemical burns (alkalis and acids) are, however, the most frequent cause of limbal ischemia and epithelial destruction causing the loss and/or impairment of LSCs function, and are the main indication for cell-based therapy approaches [18,19,20,21,22,23,24].
Figure 2. Clinical findings related to Limbal Stem Cell Deficiency (LSCD). Limbal deficiency secondary to ocular cicatricial pemphigoid with the presence of peripheral newly formed vessels leading to a loss of corneal transparency (a); Limbal deficiency, secondary to a chemical burn (bleach) of the ocular surface leads to a corneal conjuntivalization and neovascularization with loss of transparency (b). LSCD can be treated with cell therapy techniques such as cultured limbal epithelial transplantation (CLET) or cultured oral mucosal epithelial transplantation (COMET).
Figure 2. Clinical findings related to Limbal Stem Cell Deficiency (LSCD). Limbal deficiency secondary to ocular cicatricial pemphigoid with the presence of peripheral newly formed vessels leading to a loss of corneal transparency (a); Limbal deficiency, secondary to a chemical burn (bleach) of the ocular surface leads to a corneal conjuntivalization and neovascularization with loss of transparency (b). LSCD can be treated with cell therapy techniques such as cultured limbal epithelial transplantation (CLET) or cultured oral mucosal epithelial transplantation (COMET).
Jcm 04 00318 g002
Table 1. Main etiologies and pathological conditions for primary and secondary Limbal Stem Cell Deficiency (LSCD).
Table 1. Main etiologies and pathological conditions for primary and secondary Limbal Stem Cell Deficiency (LSCD).
EtiologyOcular Pathology
Idiopathic-
HereditaryAniridia
Autosomal dominant keratitis
Gelatinous drop-like corneal dystrophy
Iris coloboma
Xeroderma pigmentosa
Epidermolysis bullosa
Dyskeratosis congenita
Ectodermic dysplasia
Multiple endocrine neoplasia
Polyglandular autoimmune syndromes
NeoplasicIntraepithelial neoplasia
Conjuntival tumors (melanoma)
Limbal dermoid
DegenerativeRecurrent pterygium
Salzmann nodular corneal dystrophy
InfectionsSevere infeccious keratitis
Chlamydia conjunctivitis
MechanicalAlkali, acid, thermal burns
Bullous keratopathy
Tumor excision
Cryotherapy, radioterapy
Systemic and local chemotherapy (MMC, 5FU)
UV radition
Phototherapeutic keratectomy
AnoxicContact lenses misuse or prolonged use
TrophicNeurotrophic keratopathy
InflammationSuperior limbic keratoconjunctivitis
Collagen diseases related ulcers
Mooren ulcer
Atopic keratoconjunctivitis
Ocular pemphigoid
Ocular rosacea
Stevens-Johnson syndrome
Graft-versus-host disease
Vitamin A deficiency
MMC, mitomycin-C; 5FU, 5-fluorouracil; UV, ultra-violet.

2.1. Cell-Based Treatments for LSCD

The concept of ocular surface reconstruction was introduced with the application of autologous conjunctiva for unilateral ocular chemical alkali burns [25]. Since then, several surgical approaches have been developed with the aim of restoring the corneal epithelium on the diseased ocular surface. In recent decades, limbal transplantation techniques using auto or allografts have been introduced as bio-replacement approaches for limbal tissues to improve and reconstruct the altered ocular surface [5]. Building on previous experience treating patients with large surface areas of burned skin, the epithelial cells of the ocular surface have been obtained by cell culture techniques for ex vivo expansion. Subsequently, the ocular surface was successfully reconstructed by using LSCs in patients with severe unilateral ocular surface pathology [17]. Since then, various translational approaches have been developed and optimized, with satisfactory long-term clinical results [18,19,20,21,22,23].
Treatment approaches for LSCD can be divided into three main categories [4,5,23,24]: (a) transplants and bio-replacement of tissue; (b) cell-based therapy by ex vivo cell culture expansion; and (c) symptomatic and alternative treatment: keratoprosthesis implantation, provisional debridement of conjunctival corneal tissue, therapeutic contact lenses and drug (steroids, anti-angiogenic drugs, tear substitutes, autologous serum) therapy [5].
Ex vivo expansion of LSCs is the most innovative approach for ocular surface bio-replacement (CLET: cultured limbal epithelial transplantation). From a minimally invasive biopsy (1–2 mm2) of the healthy limbal region (the same or the contralateral eye), an explant culture technique can be applied on a suitable substrate (such as the amniotic membrane) or by separating the epithelial layer from the fragment obtained by enzymatic treatment [19,21,22,26,27,28,29]. In the latter approach, the cells obtained are in vitro co-cultured on feeder-layers (3T3 murine fibroblasts growth arrested by irradiation or mitomycin-C). Once cell growth is achieved, the cell suspensions are transferred to suitable substrates, such as fibrin, collagen or biocompatible polymers. The bio-replacement is carried out after removal of most of the diseased tissue from the ocular surface [17,18,19,20,21]. This methodology has many advantages over the tissue transplantation techniques used to date: essentially, it requires a substantially smaller limbal biopsy, which reduces the risk of limbal deficiency in healthy donor tissue. Its other advantages include a final high cell population that is more efficiently selected, homogeneous and, theoretically, more enriched with progenitor characteristic cells [26,27,28,29]. However, enzymatic techniques involve a more complex approach, with additional manipulation of the tissue and the need for xenoproducts at different stages of cell culture production. For its part, the explant technique has certain advantages—among them its technical simplicity, the lack of xenoproducts and its cost-effectiveness, despite the heterogeneity of the cell population cultured (sclera fibroblasts, antigen presenting cells, melanocytes, conjunctival epithelium cells and others) [21,22,26,27,28,29]. It is always desirable to use autologous cells for ex vivo expansion to avoid the risk of immune response. However, in the presence of severe bilateral ocular pathology, the use of heterologous epithelial cells (from cadaveric or related living donor corneas) is acceptable [18,19,20,21]. Autologous oral mucosal epithelia expanded ex vivo have also been successfully used as an alternative source of epithelial cells (COMET: cultured oral mucosal epithelial transplantation) [30,31].
The mechanism by which cultured LSCs may restore the ocular surface is still poorly understood. Cells may replace the progenitor population, and/or “reactivate” nonfunctioning host progenitor cells by providing stimuli for growth, or change niche behavior. It has been speculated that there may be “dormant” stem cells despite clinical features of LSCD [11].
Currently, ex vivo expansion methods applied in cell-based therapy for clinical application are based mostly on the use of xenogeneic or allogeneic products such as murine cells for feeder layer approaches, fetal calf or bovine serum in culture media, supplements of non-human origin for cell growth and maintenance and the human amniotic membrane as a cell carrier. These products potentially carry a risk for transmitting diseases; they may induce tumorigenesis or precipitate an immunological response in the host [18,20,21,22,23,28,29]. They also show idiosyncratic biological variability that may adversely affect the quality of cultured grafts and also the final results after transplantation. Thus, there is currently a special need to investigate options for replacing potentially hazardous xenobiotic materials with others of human origin or xeno-free chemically defined media.

2.2. Alternative Cell Sources for LSCD Treatment

Corneal transplantation (penetrating keratoplasty) is considered the conventional therapy to restore the corneal tissue. However, this technique is not a viable strategy for patients suffering LSCD because it does not replace the LSCs population [32]. Cell-based therapy is the most rational approach for ocular surface bio-replacement, and the ideal cells for corneal reconstruction are autologous corneal LSCs using CLET approach. Minimally invasive biopsy of the limbal tissue from the same patient’s healthy eye (unilateral disease) is the preferred method, although this source of progenitor cells is not always available. If both eyes present serious surface damage, the source of healthy LSCs will be lost; COMET is among the current therapeutic alternatives. In fact, therapy for LSCD is also rapidly evolving to include alternative cell types (of autologous or heterologous origin) and clinical approaches as treatment modalities. As a consequence, other strategies, such as the use of mesenchymal stem cells from adult tissue (bone marrow mesenchymal stromal cells or adipose derived stromal cells, among others) for cell regenerative therapy in corneal injuries, are gaining prominence at present. Other sources of cells or stem cells have been tested with regenerative aims in the ocular surface, and may be useful in situations where both eyes are affected although many of them, still without clinical use at present, but which have great translational potential (Table 2).
Table 2. Cell sources for ex vivo expansion cell-based therapy to treat Limbal Stem Cell Deficiency (LSCD).
Table 2. Cell sources for ex vivo expansion cell-based therapy to treat Limbal Stem Cell Deficiency (LSCD).
Cell SourcesApplicationReferences
Cultured Limbal Epithelial Cells (CLET)Clinical application[17,18,19,20,21]
Cultured Oral Mucosal Epithelial Cells (COMET)Clinical application[30,31,33,34,35,36,37]
Cultured Conjunctival Epithelial CellsClinical application[38,39,40,41]
Cultured Embryonic Stem CellsMice model[42,43,44,45]
Cultured Adult Epidermal Stem CellsGoat model[46,47,48]
Cultured Bone-Marrow Derived Mesenchymal Stem CellsRat and rabbit models[49,50,51,52,53]
Cultured Adipose Derived Mesenchymal Stem CellsIn vitro model[54,55]
Cultured Orbital Fat Mesenchymal Progenitor CellsMice model; in vitro model[56,57,58]
Cultured Immature Dental Pulp Stem CellsRabbit model[59,60]
Cultured Hair Follicle-Derived Stem CellsMice model[61,62]
Cultured Umbilical Cord Stem CellsRabbit model[63,64]
Nevertheless, the application of CLET using human amniotic membrane (hAM) or fibrin gel as a scaffold has been clinically validated and today is the most frequently used cell-based therapy applied at clinical level in ophthalmology [18,19,20,21]. Since its introduction [17], it has been used with long-term clinical follow-up periods. Despite many differences between studies regarding inclusion/exclusion criteria, the culture methods applied, transplantation techniques, and clinical outcome measures, the overall success rate of this procedure is around 70% [18,21,23]. On the other hand, oral mucosa has also been shown to be an attractive autologous epithelial cell source for cases of severe bilateral LSCD. COMET has already been used in clinical settings, offering promising long-term results with improved vision in over half of treated patients [30,31,32,33,34,35]. However, peripheral corneal neovascularization is commonly found with this approach since oral mucosal cells have greater angiogenic potential than limbal epithelial cells [65,66]. It has been suggested that these new-formed vessels may regress following local anti-angiogenic therapy [33,34,35]. Further studies are needed in this regard to assess the long-term efficacy of COMET technique. In the past five years, several clinical trials have been conducted to test, compare or consolidate the application of other approaches and other sources of progenitor cells for the treatment of LSCD (Table 3).
Table 3. Current cell-based therapy clinical trials for the treatment of Limbal Stem Cell Deficiency (LSCD).
Table 3. Current cell-based therapy clinical trials for the treatment of Limbal Stem Cell Deficiency (LSCD).
Clinical TrialIdentifierPhaseStudy CharacteristicsCell SourceSituation
Corneal Epithelium Repair and Therapy Using Autologous Limbal Stem Cell Transplantation.NCT02148016Phase 1, Phase 2Open label, Interventional Non-randomized, SGAAutologous LSCsCurrently recruiting
Multicenter Study of CAOMECS Transplantation to Patients With Total Limbal Stem Cell Deficiency.NCT01489501Phase 3Open label, Interventional Non-randomized, SGAAutologous OMCNot yet open
The Improvement of Limbal Epithelial Culture Technique by Using Collagenase to Isolate Limbal Stem Cells.NCT02202642Phase 1Open label, Interventional Non-randomized, SGAAutologous LSCsCurrently recruiting
Autologous Transplantation of Cultivated Limbal Stem Cells on Amniotic Membrane in Limbal Stem Cell Deficiency (LSD) Patients.NCT00736307Phase 1, Phase 2Open label, Interventional Non-randomized, SGAAutologous LSCsCompleted
Clinical Trial on the Effect of Autologous Oral Mucosal Epithelial Sheet Transplantation.NCT02149732Phase 1, Phase 2Open label, Interventional Non-randomized, SGAAutologous OMECCurrently recruiting
Cultivated Stem Cell Transplantation for the Treatment of Limbal Stem Cell Deficiency (LECT).NCT00845117Phase 1, Phase 2Open label, Interventional Non-randomized, SGAAutologous LSCsOngoing, but not recruiting
Limbal Epithelial Stem Cell Transplantation: a Phase II Multicenter Trial (MLEC)NCT02318485Phase 2Open label, Interventional Non-randomized, SGAAllogenic or autologous LSCsNot yet open
Cell Therapy in Failure Syndromes in Limbal Stem Cells (TC181).NCT01619189Phase 2Single blind, Interventional Non-randomized, SGAAllogenic or autologous LSCsCurrently recruiting
Autologous Cultured Corneal Epithelium (CECA) for the Treatment of Limbal Stem Cell Deficiency.NCT01756365Phase 1, Phase 2Open label, Interventional Non-randomized, SGAAutologous cultured corneal epitheliumEnrolling by invitation
Ocular Surface Reconstruction With Cultivated Autologus Mucosal Epithelial Transplantation.NCT01942421Phase 2, Phase 3Open label, Interventional Non-randomized, SGAAutologous OMECOngoing, but not recruiting
Efficacy of Cultivated Corneal Epithelial Stem Cell for Ocular Surface Reconstruction.NCT01237600Phase 2, Phase 3Open label, Interventional Non-randomized, SGAAllogenic or autologous LSCsCompleted
Safety Study of Stem Cell Transplant to Treat Limbus Insufficiency Syndrome.NCT01562002Phase 1, Phase 2Double blind, Interventional Randomized, Parallel assignmentAllogenic LSCs vs. BM-MSCsOngoing, but not recruiting
The Application of Oral Mucosal Epithelial Cell Sheets Cultivated on Amino Membrane in Patients Suffering From Corneal Stem Cell Insufficiency or Symblepharon.NCT00491959Phase 1Open label, Interventional Non-randomized, SGAAutologous OMECCompleted
Transplantation of Cultivated Corneal Epithelial Sheet in Patients With Ocular Surface Disease (CLET).NCT01123044Phase 3Open label, Interventional Randomized, Parallel assignmentAutologous LSCsUnknown
Application of Cell Therapy for Ocular Surface Repair Using Progenitor Cells of Sclerocorneal Limbus Amplified ex vivo (MeRSO09).NCT01470573Phase 2Open label, Interventional Non-randomized, SGAAutologous LSCsCompleted
The Application of Cultured Cornea Stem Cells in Patients Suffering From Corneal Stem Cell Insufficiency.NCT01377311Phase 1Open label, Interventional Non-randomized, SGAAutologous LSCsCompleted
LSCs, limbal stem cells; OMEC, oral mucosal epithelial cells; BM-MSCs, bone marrow derived mesenchymal stem cells; SGA, single group assignment [67].

3. IPSCs and Corneal Epithelial Differentiation

As discussed above, adult stem cells make it possible to repair and regenerate damaged epithelial tissue. In general, these cells reside in the basal layer of the epithelium, are able to self-renew continuously, and produce transient amplifying cells (TACs) that differentiate terminally after a brief period of proliferation [68,69,70,71,72]. However, there are limitations to LSCs transplantation therapies. On one hand, for unilateral LSCD, taking biopsies from the healthy eye carries along the risk of damaging the donor eye. On the other hand, for bilateral LSCD, allogenic transplantation presents the risk of immune rejection by the patient. In this sense, induced pluripotent stem cells (IPSCs) can be obtained from minimally invasive sources from the patient himself and be differentiated into LSCs, avoiding immune rejection problems and cell availability. The discovery of IPSCs has been one of the most significant advances in regenerative medicine in the last decade. Overexpression of a specific set of transcription factors (e.g., Oct4, Sox2, c-Myc, and Klf4; or Oct4, Sox2, Lin28 and Nanog) in adult differentiated cells can reprogram cell fate and IPSCs [68,69,70]. These can be differentiated into various cell types, a property that has opened up a wide range of possibilities for the investigation of cell states, the mechanisms of differentiation, pluripotency and other related cellular identities and behaviors. Contrary to embryonic stem cells, IPSCs can be created from easy access differentiated cells, such as fibroblast or keratinocytes, and allow the creation of autologous sources of different cell types for regenerative therapies or disease modeling.
More recently, direct reprogramming of cells into different states (either pluripotent or somatic) offers one of the most promising approaches in the field of regenerative medicine, with enormous potential for examining clinical and therapeutic applications in more depth [71]. The “direct reprogramming” is characterized by a process wherein mature, fully differentiated somatic cells, can be induced to other cell types without necessarily going through a pluripotent state [71]. To this end, cells can be reprogrammed by transient overexpression of transcriptional factors for a relatively short time interval. The cells in this state are called IPS-partial cells; they respond to different signal environments (e.g., growth factors, cytokines, inductors agents, etc.) and have the ability to direct cell fate decisions in reprogramming [71]. For corneal repair direct reprogramming would be of great advantage by not only eliminating the pluripotent stages (potentially carcinogenic) but also avoiding the lengthy production and characterization of IPSC lines. As very few cells are needed for ocular surface cell therapy, the limited expansion capacity of IPSCs is not a limiting factor as well as the production time, which would be much shorter with an easier methodology. However, there are still very limited references to the LSCs production by transdifferentiation from easily accessible adult somatic cells. Rat adult stem cells from the bulge of hair follicle were transdifferentiated into corneal epithelial-like cells by culturing with corneal limbus soluble factors and forced overexpression of the transcripton factor Pax6 [73]. More recently, Sainchanma and colleagues [74] described a method to obtain corneal epithelial-like cells from human skin-derived precursor cells—which present some multipotency markers—by culturing them with three specific growth factors: epidermal growth factor (EGF), keratinocyte growth factor (KGF) and hepatocyte growth factor (HGF) [74]. These are encouraging results to open the way for new sources of LSCs autologous supply. However further work is necessary to refine the protocol to obtain final cells that are closer to LSCs in their marker profiling and their functionality in restoring corneal epithelium should be tested.

3.1. Application of IPSCs for Ocular Pathology

Regarding the application of IPSCs in the field of cell therapy for ocular pathology, IPSCs have shown great promise in treating certain degenerative retinal diseases, particularly those that affect the functionality of the retinal pigment epithelium (RPE) due to its dysfunction or loss. In this context—dry age-related macular degeneration (geographic atrophy)—cell-based therapy may be a rational and effective therapeutic alternative for certain forms of retinitis pigmentosa and gyrate atrophy [75].
The use of stem cell therapy for eye diseases presents many advantages, for a variety of reasons: (a) the intraocular environment benefits from a state of immune privilege; (b) the target tissue to be treated has certain individual anatomical and functional characteristics (defined subretinal space and specialized single stratified epithelium); (c) the intraocular space is small and limited, as is required by the treatment given the low number of cells involved; and (d) the intraocular space is easily controllable by sophisticated diagnostic imaging systems in ophthalmology that allow convenient monitoring with satisfactory clinical follow up—for example, by injection of cells under the subretinal space or into the vitreous body—which permits the visualization of the therapeutic effect and possible complications. For this reason, several recent research studies are being carried out [75,76,77].
The regeneration of the ocular surface and restoration of corneal transparency following injury is one of the fields where IPSCs may also be applicable. The first attempt to obtain LSC-like cells from pluripotent cells were carried out by Notara and colleagues [78]. Using mouse ESCs treated with conditioned media from limbal fibroblasts they obtained cells with cobblestone morphology that expressed cytokeratin (CK) 12 and ΔNp63α, opening the door for the study of pluripotent cells-derived cells in the regeneration of corneal epithelium. Yu and co-researchers [79] also obtained about 13% of conversion of mouse IPSCs to corneal epithelium-like cells by co-culture of IPSCs with corneal limbal stroma in the presence of additional growth factors related to corneal development: basic fibroblast growth factor (bFGF), EGF and nerve growth factor (NGF). Moving toward human cells, Hayashi and colleagues [80] aimed to establish IPSCs derived from human LSCs and to examine the ability of both limbal-derived and human dermal fibroblast-derived IPSCs to differentiate into corneal epithelial cells. Corneal epithelial cells were then successfully induced by the stromal cell-derived inducing activity (SDIA) differentiation method, after prolonged differentiation culture (12 weeks or later) in both, limbal (with higher corneal epithelial differentiation efficiency) and fibroblastic IPSCs. This study was the first to demonstrate a strategy for corneal epithelial cell differentiation from human IPSCs, and further suggested that an epigenomic status related to DNA methylation in specific epithelium-related genes—CK3, CK12 and Pax6—was associated with the propensity of IPSCs to differentiate into corneal epithelial cells and could be used as a criteria to choose IPSCs source for LSCs differentiation However, this protocol is lengthy and the efficiency is low, as the population obtained after the differentiation protocol is mixed with other cell types, such as RPE or lens epithelium. Ljubimov’s group [81] recently successfully generated IPSCs from human primary LSCs to re-differentiate these IPSCs back into the limbal corneal epithelium, maintaining them on natural substrate that mimicked the native LSC niche, including denuded hAM and de-epithelialized corneas. This choice of parent cells represented an improvement for limbal cell differentiation by partial retention of parental epigenetic signatures in IPSCs. The authors observed that when the gene methylation patterns were compared in IPSCs to parental LSCs, limbal-derived IPSCs presented fewer unique methylation changes than fibroblast-derived IPSCs, suggesting the retention of epigenetic memory (genes promoting methylation) during reprogramming. Interestingly, limbal-derived IPSCs cultured for two weeks on hAM induced markedly higher expression of LSC markers (ABCG2, ΔNp63, CK14, CK15, CK17, N-cadherin, and TrkA) than fibroblast-derived IPSCs. On hAM, the methylation profiles of select limbal-derived IPSC genes became closer to the parental cells, but fibroblast-derived IPSCs remained closer to parental fibroblasts. On denuded air-lifted corneas, limbal-derived IPSCs even upregulated differentiated corneal CK3 and CK12. Taking all the data together, the authors emphasize the importance of the natural niche and the limbal tissue of origin in generating IPSCs as LSCs for clinical aims [81]. Compared to the previous work of Hayashi and colleagues [80], this method presents two interesting improvements. The differentiation medium is serum free and contains defined growth supplements, allowing for a more standardized protocol and bringing it closer to a clinical application. Also, differentiating the IPSCs on hAM provides an advantage for the success of future transplantation [21]. Both research [80,81] lead to the conclusion that the initial cell type from which IPSCs are derived is important for the quality of the final LSC-like cells obtained. However, for clinical applications easily accessible donor cell types should be identified to create IPSCs-LSCs. Other than fibroblasts, adult progenitor cells like bone-marrow or hair follicle -derived mesenchymal stem cells should be also tested.
In an elegant approach applying a directed two-stage differentiation protocol without the use of feeder cells or serum in the culture medium, researchers generated relatively pure populations of corneal epithelial-like progenitor cells capable of terminal differentiation toward mature corneal epithelial-like cells [82]. Early developmental mechanisms could be reproduced in vitro by blocking the transforming growth factor β (TGF-β) and Wnt-signaling pathways with small molecule inhibitors and activating bFGF signaling. IPSCs were cultured onto collagen IV substrate in specific corneal epithelial cell growth media which differentiated them into LSCs. Cells expressed typical LSC markers such as cytokeratins (CK3, CK12 and CK15) as well as Pax6, ABCG2 and ΔNp63 after five weeks of differentiation [82]. Interestingly, the differentiation protocol described by the authors, using growth factors and small molecules inhibitors, can be performed totally in xeno-free, feeder-free and serum-free conditions, allowing for a reproducible and clinical grade production of the IPSCs-LSCs ready to be used in the clinical setting. To bring one more step closer into the clinic, Wu’s team [83] described a IPSCs-LSCs transplantation system that introduces a 3D scaffolding in which IPSCs are seeded, differentiated and grafted into an acellular porcine matrix scaffold. This bioengineering system is aimed to overcome the gradual loss of viability over time of LSC grafted cells and the limitations of amniotic membrane. The method improved the outcome in rabbit experimental models [83].
In conclusion, even if several adult stem cells types have been used for regeneration of corneal epithelium, LSCs themselves have shown superior results and are the cells of choice for LSCD treatment. Since IPSCs grow indefinitely, IPSC-derived LSCs are an unlimited source of autologous LSCs for patients with bilateral LSCD – and therefore no LSCs left - and to avoid the risks of surgical intervention in unilateral LSCD. Moreover, the idea of creating IPSCs banks to provide HLA matched (immune-compatible) tissues is being strongly considered by the scientific community [84]. This would provide a ready-to-go source of material for LSCs derivation avoiding the high cost of personalized IPSCs development.

3.2. Molecular Mechanisms of Corneal Epithelial Reprogramming

Molecular mechanisms of epithelial reprogramming have been analyzed, and ΔNp63 has emerged as a central protein in IPSC reprogramming routes. ΔNp63 has been found to enhance IPSCs generating efficiency, as the loss of function of this protein decreased the mesenchymal-epithelial transition (MET) and pluripotency genes [85]. Accordingly, in cell oncogenic transformation (a process that may share similarities with IPSCs reprogramming at signaling level) it was found that Oct4 upregulation could enhance the expression of ΔNp63 while repressing p53 [86]. APR-246/PRIMA-1met (a small compound which restores the functionality of mutant p53 in human tumor cells that target mutant forms of ΔNp63) was found to reverse corneal epithelial lineage commitment and to reinstate a normal p63-related signaling pathway [87]. In this study [87], the authors designed a unique cellular model that recapitulated major embryonic defects related to ectrodactyly ectodermal dysplasia cleft (lip/palate) syndrome (EEC syndrome), which is caused by single point mutations in the p63 gene. Fibroblasts from healthy donors and from EEC patients carrying two different point mutations in the DNA binding domain of p63 were reprogrammed into IPSC lines. Phenotypic defects in EEC syndrome include skin defects and LSCD, with loss of corneal transparency. In this interesting in vitro model, EEC-derived IPSCs failed to terminal differentiate into CK14 cells (epidermis and LSCs) or CK3/CK12 cells (corneal epithelial cells) [87]. This research team also described previously the possible roles of specific miRs in corneal development using IPSC corneal differentiation methods [88]. Similarly, IPSC epithelial somatic differentiation seems to recapitulate the molecular steps during embryonic development, in which ΔNp63 is a master regulator of epithelial differentiation. Moreover, during IPSCs generation it is widely accepted that MET is needed [89]. Blocking MET during cell reprogramming (using TGF-β or Snail1) prevents IPSCs induction. In this change of cell state, the inverse of MET occurs during embryonic development, in which epithelial-mesenchymal transition (EMT) pointing out the parallelism between embryonic development and cell reprogramming [89]. Another interesting signaling pathway that may be involved in the IPSC differentiation into epithelial cells is the Pax6/β-catenin pathway [90]. During the embryonic development of the chicken eye, eye specification seems to be established by the inhibition of the canonical Wnt pathway and TGF-β, which induces the upregulation of Pax6 in the lens ectoderm [87]. In support of this theory, the trans-differentiation of multipotent hair follicle stem cells into corneal epithelial-like cells is mediated by the upregulation of Pax6 and the inhibition of the canonical Wnt-signaling pathway [73]. Thus, further investigation is needed to clarify whether this mechanism really affects the differentiation of IPSCs into corneal epithelial cells.

3.3. Restoration of Corneal Stromal Transparency

The restoration of corneal transparency after stromal or endothelial damage is another field of interest in which IPSCs generation and differentiation may have an impact. The production of corneal keratocytes from pluripotent cells also has significant implications for cell-based therapy and tissue engineering for treatment of corneal diseases. At present, however, there are very few studies of the use of IPSCs as an effective and conclusive approach for cell therapy applications for recovery corneal stroma, and the results are very preliminary.
Funderburgh’s group [91] developed a methodology for inducing the differentiation of human embryonic stem cells (hESCs) into cells with a gene-expression phenotype similar to that of adult human corneal keratocytes. The transparency of the cornea depends on the unique molecular composition and organization of the extracellular matrix of the stroma (collagen fibrils), which is a product of keratocytes—specialized neural crest (NC)-derived mesenchymal cells. In Funderburgh’s study, neural differentiation of the hESC cell line was induced by co-culture with mouse PA6 fibroblasts as a feeder-layer. After a few days in co-culture, hESCs acquired the ability to express cell-surface nerve growth factor receptor (NGFR, p75NTR) of low affinity. These cells were then isolated from co-cultures by immunoaffinity adsorption and cultured further as a monolayer. Corneal keratocyte phenotype was induced in serum-free medium containing ascorbate and was independent of the substratum for cultivation. Interestingly, hESC co-cultures upregulated the expression of some specific NC genes, and when NGFR-expressing cells were expanded as a monolayer, mRNAs typifying adult stromal stem cells were detected. Further, when these cells were cultured as substratum-free pellets, several corneal keratocyte markers were upregulated, among them keratocan, a corneal stroma-specific proteoglycan. The analysis of culture medium obtained from the pellets also contained high concentrations of keratocan modified with keratan sulfate, considered a unique molecular component of corneal stroma. This study showed the possibility to differentiate keratocytes in vitro. The authors also hypothesized that IPSCs derived from adult somatic cells could be used in place of hESCs for both, to provide autologous material for bioengineered corneal matrix or for direct stromal cell-based therapy [91].
Human corneal keratocytes could also be reprogrammed into IPSCs exhibiting pluripotent properties. To prevent feeder cell contamination and to improve the clinical utility of reprogrammed IPSCs, Chien and colleagues [92] developed a feeder-free (without MEF, mouse embryonic fibroblasts cells) and serum-free method to stably expand human IPSCs in vitro. This approach allows cells to remain stable through 30 passages, maintaining ESC-like pluripotent properties. Furthermore, to improve IPSCs delivery and engraftment, a biocompatible injectable nanogel (thermo-gelling carboxymethyl-hexanoyl chitosan; CHC) was developed. The authors also evaluated whether the viability and pluripotent properties of human corneal keratocyte-derived IPSCs can be retained in a CHC hydrogel system, and explored the therapeutic potential of these cells on corneal impairment using CHC hydrogel as delivery vehicle in a rat model of corneal damage induced by either chemical burns or surgical ablation. They concluded that the IPSC/CHC system enhanced corneal regeneration by downregulating oxidative stress and recruiting endogenous epithelial cells to restore corneal epithelial thickness, and also reconstructing the corneal microenvironment niche [92].
Very recently, Fukuta and co-researchers [93] developed an efficient induction protocol using chemically defined culture medium containing inhibitors for TGF-β signaling and inhibitors for Wnt-signaling pathway (GSK3β). This approach allow differentiate human neural crest cells (hNCC) from human pluripotent cells, with the same efficiency (70%–80%), independent of the parental cell type (ESCs or IPSCs), or method of generation (viral-integrated or plasmid-episomal). Furthermore, cells have been kept under feeder-free and xeno-free culture systems. Interestingly, generated hNCCs could be differentiated into corneal endothelial cells, among other complex cell types, such as peripheral neurons, glial cells and melanocytes. Endothelial cells of the cornea have been differentiated culturing hNCCs in corneal endothelial cell conditioned medium supplemented with selective ROCK inhibitor (Y-27632). After two weeks of induction, cells changed their morphology into that of polygonal corneal endothelial-like cells and started to express ZO-1, type IV and type VIII collagens, which are recognized corneal endothelial cell markers [93]. These results also open new and promising perspectives for possible clinic applications in corneal pathologies where the endothelium is primarily affected.

3.4. Future Trends for IPSC Technology

The ideal source of cells for ocular clinical application needs to meet certain criteria: (a) easy accessibility and minimal risks for patients; (b) availability in sufficient quantities for bio-replacement; and (c) a high likelihood of successful reprogramming [72]. However, present evidence confirms that the methods involving IPSCs production should be considered with caution before immediate clinical application. An example is exome sequencing of several human IPSC lines, identified over a hundred point mutations in the generated cells but not in the parental cells. Many missense mutations associated with the function of different proteins and other point mutations in genes related to cancers have been observed [94]. In this sense, a better understanding of the molecular mechanisms for differentiation into various cell types associated with more directed protocols for the reprogramming, without the need to induce complete states of non-differentiation, could contribute to mitigate possible aberrations in the genome of produced cell populations.

4. Conclusions

In recent years there have been significant developments in the use of cell-based cultures combined with biomaterials or biocompatible substrates for corneal epithelial tissue engineering bio-replacement. Current approaches for improving these therapeutic strategies include standardization of culture conditions and development of xenobiotic free culture systems, evaluation of novel bio-functional scaffolds to enhance stem cell expansion and transplantation efficacy, and exploration of alternative autologous progenitor cell sources. To date, the search for innovative strategies and approaches in the field of ocular surface reconstruction has produced some encouraging results. Several new strategies have emerged for future therapies for LSCD, although the best cell source and the ideal technique still need to be established. One of the key elements is the role of the cellular microenvironment or niche. The limbal stem cell niche contains stem cells that promote proliferation and migration and have immunosuppressive mechanisms to protect them from immunological reactions. The current findings suggest that the CLET and COMET approaches using autologous epithelial progenitor cells are the most widely accepted clinical techniques for treating LSCD.
One emerging alternative cell source for treating the ocular surface is the use of adult stem cells, which provide high proliferative potential, differentiated capability and lower immunogenicity; they are non-tumorigenic and can be obtained by minimally invasive methodologies. They represent a more physiological, more rational, and less invasive treatment. Meanwhile, stem cells from adult tissue, as in the case of mesenchymal stem cells, although they have showed an intrinsic potential for a possible epithelial differentiation, this has not yet been achieved. Also, the prospects for therapies derived from autologous mesenchymal and IPSCs that may yield a multitude of engineered tissue types are exciting. Although IPSCs are yet to be used for ocular surface reconstruction, a recent study has shown successful corneal epithelial cell generation. The search for alternative sources of stem cells in the treatment of ocular surface diseases represents a challenge. IPSCs represent a very promising option for obtaining corneal epithelial cells to apply in cell-based therapy for the ocular surface. In the future, a deeper understanding of the behavioral characteristics of the LSC niche as well as of proliferation and differentiation pathway events should help to expand and develop the use of IPSCs in ocular surface regenerative medicine.

Acknowledgements

Part of this work was funded by Fondo de Investigaciones Sanitarias del Instituto de Salud Carlos III (FIS09-PI040654), Dirección General de Terapias Avanzadas y Trasplantes del Ministerio de Sanidad y Política Social (TRA-072), and Fundació Marató TV3 (120630).

Author Contributions

Núria Nieto-Nicolau and Eva M. Martínez-Conesa performed the literature searches and bibliography, and contributed to the writing of the manuscript. Michael Edel contributed to draft the manuscript. Ana B. Álvarez-Palomo contributed to perform literature searches and bibliography compilations, to draft and revise the final manuscript. Ricardo P. Casaroli-Marano drafted, formatted and wrote the manuscript, prepared the figures and tables, performed literature searches and bibliography compilations, and edited and revised the final manuscript.

Conflicts of Interest

The authors have no proprietary or financial interest in any materials, methods or subject matter discussed in this article.

References

  1. Notara, M.; Alatza, A.; Gilfillan, J.; Harris, A.R.; Levis, H.J.; Schrader, S.; Vernon, A.; Daniels, J.T. In sickness and in health: Corneal epithelial stem cell biology, pathology and therapy. Exp. Eye Res. 2010, 90, 188–195. [Google Scholar] [CrossRef] [PubMed]
  2. Osei-Bempong, C.; Figueiredo, F.C.; Lako, M. The limbal epithelium of the eye—A review of limbal stem cell biology, disease and treatment. Bioessays 2013, 35, 211–219. [Google Scholar] [CrossRef] [PubMed]
  3. Casaroli-Marano, R.P.; Nieto-Nicolau, N.; Martínez-Conesa, E.M. Progenitor cells for ocular surface regenerative therapy. Ophthalmic Res. 2013, 49, 115–121. [Google Scholar] [CrossRef] [PubMed]
  4. Ahmad, S.; Kolli, S.; Lako, M.; Figueiredo, F.C.; Daniels, J.T. Stem cell therapies for ocular surface disease. Drug Discov. Today 2010, 15, 306–313. [Google Scholar] [CrossRef] [PubMed]
  5. Utheim, T.P. Limbal epithelial cell therapy: Past, present, and future. Methods Mol. Biol. 2013, 1014, 3–43. [Google Scholar] [PubMed]
  6. Li, W.; Hayashida, Y.; Chen, Y.T.; Tseng, S.C. Niche regulation of corneal epithelial stem cells at the limbus. Cell Res. 2007, 17, 26–36. [Google Scholar] [CrossRef] [PubMed]
  7. Dua, H.S.; Shanmuganathan, V.A.; Powell-Richards, A.O.; Tighe, P.J.; Joseph, A. Limbal epithelial crypts: A novel anatomical structure and a putative limbal stem cell niche. Br. J. Ophthalmol. 2005, 89, 529–532. [Google Scholar] [CrossRef] [PubMed]
  8. Shortt, A.J.; Secker, G.A.; Munro, P.M.; Khaw, P.T.; Tuft, S.J.; Daniels, J.T. Characterization of the limbal epithelial stem cell niche: Novel imaging techniques permit in vivo observation and targeted biopsy of limbal epithelial stem cells. Stem Cells 2007, 25, 1402–1409. [Google Scholar] [CrossRef] [PubMed]
  9. Thoft, R.A.; Friend, J. The X, Y, Z hypothesis of corneal epithelial maintenance. Invest. Ophthalmol. Vis. Sci. 1987, 24, 1442–1443. [Google Scholar]
  10. Majo, F.; Rochat, A.; Nicolas, M.; Jaoude, G.A.; Barrandon, Y. Oligopotent stem cells are distributed throughout the mammalian ocular surface. Nature 2008, 456, 250–254. [Google Scholar] [CrossRef] [PubMed]
  11. Dua, H.S.; Miri, A.; Alomar, T.; Yeung, A.M.; Said, D.G. The role of limbal stem cells in corneal epithelial maintenance: Testing the dogma. Ophthalmology 2009, 116, 856–863. [Google Scholar] [CrossRef] [PubMed]
  12. 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. [Google Scholar] [CrossRef] [PubMed]
  13. Lindberg, K.; Brown, M.E.; Chaves, H.V.; Kenyon, K.R.; Rheinwald, J.G. In vitro propagation of human ocular surface epithelial cells for transplantation. Invest. Ophthalmol. Vis. Sci. 1993, 34, 2672–2679. [Google Scholar] [PubMed]
  14. Kruse, F.E. Stem cells and corneal epithelial regeneration. Eye 1994, 8, 170–183. [Google Scholar] [CrossRef] [PubMed]
  15. Kiritoshi, A.; Sundar-Raj, N.; Thoft, R.A. Differentiation in cultured limbal epithelium as defined by keratin expression. Invest. Ophthalmol. Vis. Sci. 1991, 32, 3073–3077. [Google Scholar] [PubMed]
  16. Cotsarelis, G.; Cheng, S.Z.; Dong, G.; Sun, T.T.; Lavker, R.M. 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. [Google Scholar] [CrossRef] [PubMed]
  17. Pellegrini, G.; Traverso, C.E.; Franzi, A.T.; Zingirian, M.; Cancedda, R.; de Luca, M. Long-term restoration of damaged corneal surfaces with autologous cultivated corneal epithelium. Lancet 1997, 349, 990–993. [Google Scholar] [CrossRef] [PubMed]
  18. Shortt, A.J.; Secker, G.A.; Notara, M.D.; Limb, G.A.; Khaw, P.T.; Tuft, S.J.; Daniels, J.T. Transplatation of ex vivo cultured limbal epitehlial stem cells: A review of techniques and clinical results. Surv. Ophthalmol. 2007, 52, 483–502. [Google Scholar] [CrossRef] [PubMed]
  19. Rama, P.; Matuska, S.; Paganoni, G.; Spinelli, A.; de Luca, M.; Pellegrini, G. Limbal stem-cell therapy and long-term corneal regeneration. N. Engl. J. Med. 2010, 363, 147–155. [Google Scholar] [CrossRef] [PubMed]
  20. Sangwan, V.S.; Basu, S.; Vemuganti, G.K.; Sejpal, K.; Subramaniam, S.V.; Bandyopadhyay, S.; Krishnaiah, S.; Gaddipati, S.; Tiwari, S.; Balasubramanian, D. Clinical outcomes of xeno-free autologous cultivated limbal epithelial transplantation: A 10-year study. Br. J. Ophthalmol. 2011, 95, 1525–1529. [Google Scholar] [CrossRef] [PubMed]
  21. Baylis, O.; Figueiredo, F.; Henein, C.; Lako, M.; Ahmad, S. 13 years of cultured limbal epithelial cell therapy: A review of the outcomes. J. Cell Biochem. 2011, 112, 993–1002. [Google Scholar] [CrossRef] [PubMed]
  22. Pellegrini, G.; Rama, P.; di Rocco, A.; Panaras, A.; de Luca, M. Concise review: Hurdles in a successful example of limbal stem cell-based regenerative medicine. Stem Cells 2014, 32, 26–34. [Google Scholar] [CrossRef] [PubMed]
  23. Menzel-Severing, J.; Kruse, F.E.; Schlötzer-Schrehardt, U. Stem cell-based therapy for corneal epithelial reconstruction: Present and future. Can. J. Ophthalmol. 2013, 48, 13–21. [Google Scholar] [CrossRef] [PubMed]
  24. Ahmad, S. Concise review: Limbal stem cell deficiency, dysfunction, and distress. Stem Cells Transl. Med. 2012, 1, 110–115. [Google Scholar] [CrossRef] [PubMed]
  25. Mönks, T.; Busin, M. Modified technique of autologous conjunctival transplantation after corneal chemical burn. Klin. Monbl. Augenheilkd. 1994, 204, 121–125. [Google Scholar] [CrossRef] [PubMed]
  26. Ahmad, S.; Osei-Bempong, C.; Dana, R.; Jurkunas, U. The culture and transplantation of human limbal stem cells. J. Cell Physiol. 2010, 225, 15–19. [Google Scholar] [CrossRef] [PubMed]
  27. Tseng, S.C.; Chen, S.Y.; Shen, Y.C.; Chen, W.L.; Hu, F.R. Critical appraisal of ex vivo expansion of human limbal epitelial stem cells. Curr. Mol. Med. 2010, 10, 841–850. [Google Scholar] [CrossRef] [PubMed]
  28. Di Iorio, E.; Ferrari, S.; Fasolo, A.; Böhm, E.; Ponzin, D.; Barbaro, V. Techniques for culture and assessment of limbal stem cell grafts. Ocul. Surf. 2010, 8, 146–153. [Google Scholar] [CrossRef] [PubMed]
  29. O’Callaghan, A.R.; Daniels, J.T. Limbal epithelial stem cell therapy: Controversies and challenges. Stem Cells 2011, 29, 1923–1932. [Google Scholar] [CrossRef] [PubMed]
  30. Nishida, K.; Yamato, M.; Hayashida, Y.; Watanabe, K.; Yamamoto, K.; Adachi, E.; Nagai, S.; Kikuchi, A.; Maeda, N.; Watanabe, H.; et al. Corneal reconstruction with tissue engineered cell sheets composed of autologous oral mucosal epithelium. N. Engl. J. Med. 2004, 351, 1187–1196. [Google Scholar] [CrossRef] [PubMed]
  31. Kolli, S.; Ahmad, S.; Mudhar, H.S.; Meeny, A.; Lako, M.; Figueiredo, F.C. Successful application of ex vivo expanded human autologous oral mucosal epithelium for the treatment of total bilateral limbal stem cell deficiency. Stem Cells 2014, 32, 2135–2146. [Google Scholar] [CrossRef] [PubMed]
  32. Vemuganti, G.K.; Fatima, A.; Madhira, S.L.; Basti, S.; Sangwan, V.S. Limbal stem cells: Application in ocular biomedicine. Int. Rev. Cell Mol. Biol. 2009, 275, 133–181. [Google Scholar] [PubMed]
  33. Priya, C.G.; Arpitha, P.; Vaishali, S.; Prajna, N.V.; Usha, K.; Sheetal, K.; Muthukkaruppan, V.; et al. Adult human buccal epithelial stem cells: Identification, ex-vivo expansion, and transplantation for corneal surface reconstruction. Eye 2011, 25, 1641–1649. [Google Scholar] [CrossRef] [PubMed]
  34. Nakamura, T.; Takeda, K.; Inatomi, T.; Sotozono, C.; Kinoshita, S. Long-term results of autologous cultivated oral mucosal epithelial transplantation in the scar phase of severe ocular surface disorders. Br. J. Ophthalmol. 2011, 95, 942–946. [Google Scholar] [CrossRef] [PubMed]
  35. Liu, J.; Sheha, H.; Fu, Y.; Giegengack, M.; Tseng, S.C. Oral mucosal graft with amniotic membrane transplantation for total limbal stem cell deficiency. Am. J. Ophthalmol. 2011, 152, 739–747. [Google Scholar] [CrossRef] [PubMed]
  36. Inatomi, T.; Nakamura, T.; Kojyo, M.; Koizumi, N.; Sotozono, C.; Kinoshita, S. Ocular surface reconstruction with combination of cultivated autologous oral mucosal epithelial transplantation and penetrating keratoplasty. Am. J. Ophthalmol. 2006, 142, 757–764. [Google Scholar] [CrossRef] [PubMed]
  37. Takeda, K.; Nakamura, T.; Inatomi, T.; Sotozono, C.; Watanabe, A.; Kinoshita, S. Ocular surface reconstruction using the combination of autologous cultivated oral mucosal epithelial transplantation and eyelid surgery for severe ocular surface disease. Am. J. Ophthalmol. 2011, 152, 195–201. [Google Scholar] [CrossRef] [PubMed]
  38. Sangwan, V.S.; Vemuganti, G.K.; Iftekhar, G.; Bansal, A.K.; Rao, G.N. Use of autologous cultured limbal and conjunctival epithelium in a patient with severe bilateral ocular surface disease induced by acid injury: A case report of unique application. Cornea 2003, 22, 478–481. [Google Scholar] [CrossRef] [PubMed]
  39. Tan, D.T.; Ang, L.P.; Beuerman, R.W. Reconstruction of the ocular surface by transplantation of a serum-free derived cultivated conjunctival epithelial equivalent. Transplantation 2004, 77, 1729–1734. [Google Scholar] [CrossRef] [PubMed]
  40. Ang, L.P.; Tanioka, H.; Kawasaki, S.; Ang, L.P.; Yamasaki, K.; Do, T.P.; Thein, Z.M.; Koizumi, N.; Nakamura, T.; Yokoi, N.; et al. Cultivated human conjunctival epithelial transplantation for total limbal stem cell deficiency. Invest. Ophthalmol. Vis. Sci. 2010, 51, 758–764. [Google Scholar] [CrossRef] [PubMed]
  41. Subramaniam, S.V.; Sejpal, K.; Fatima, A.; Gaddipati, S.; Vemuganti, G.K.; Sangwan, V.S. Coculture of autologous limbal and conjunctival epithelial cells to treat severe ocular surface disorders: Long-term survival analysis. Indian J. Ophthalmol. 2013, 61, 202–207. [Google Scholar] [CrossRef] [PubMed]
  42. Homma, R.; Yoshikawa, H.; Takeno, M.; Kurokawa, M.S.; Masuda, C.; Takada, E.; Tsubota, K.; Ueno, S.; Suzuki, N. Induction of epithelial progenitors in vitro from mouse embryonic stem cells and application for reconstruction of damaged cornea in mice. Invest. Ophthalmol. Vis. Sci. 2004, 45, 4320–4326. [Google Scholar] [CrossRef] [PubMed]
  43. Ueno, H.; Kurokawa, M.S.; Kayama, M.; Homma, R.; Kumagai, Y.; Masuda, C.; Takada, E.; Tsubota, K.; Ueno, S.; Suzuki, N. Experimental transplantation of corneal epithelium-like cells induced by Pax6 gene transfection of mouse embryonic stem cells. Cornea 2007, 26, 1220–1227. [Google Scholar] [CrossRef] [PubMed]
  44. Ahmad, S.; Stewart, S.R.; Yung, S.; Kolli, S.; Armstrong, L.; Stojkovic, M.; Figueiredo, F.; Lako, M. Differentiation of human embryonic stem cells into corneal epithelial-like cells by in vitro replication of the corneal epithelial stem cell niche. Stem Cells 2007, 25, 1145–1155. [Google Scholar] [CrossRef] [PubMed]
  45. Hanson, C.; Hardarson, T.; Ellerstrom, C.; Nordberg, M.; Caisander, G.; Rao, M.; Hyllner, J.; Stenevi, U. Transplantation of human embryonic stem cells onto a partially wounded human cornea in vitro. Acta Ophthalmol. 2013, 91, 127–130. [Google Scholar] [CrossRef] [PubMed]
  46. Gao, N.; Wang, Z.; Huang, B.; Ge, J.; Lu, R.; Zhang, R.; Fan, Z.; Lu, L.; Peng, Z.; Cui, G. Putative epidermal stem cell convert into corneal epithelium-like cell under corneal tissue in vitro. Sci. China C Life Sci. 2007, 50, 101–110. [Google Scholar] [CrossRef] [PubMed]
  47. Yang, X.; Qu, L.; Wang, X.; Zhao, M.; Li, W.; Hua, J.; Shi, M.; Moldovan, N.; Wang, H.; Dou, Z. Plasticity of epidermal adult stem cells derived from adult goat ear skin. Mol. Reprod. Dev. 2007, 74, 386–396. [Google Scholar] [CrossRef] [PubMed]
  48. Yang, X.; Moldovan, N.I.; Zhao, Q.; Mi, S.; Zhou, Z.; Chen, D.; Gao, Z.; Tong, D.; Dou, Z. Reconstruction of damaged cornea by autologous transplantation of epidermal adult stem cells. Mol. Vis. 2008, 14, 1064–1070. [Google Scholar] [PubMed]
  49. Ma, Y.; Xu, Y.; Xiao, Z.; Yang, W.; Zhang, C.; Song, E.; Du, Y.; Li, L. Reconstruction of chemically burned rat corneal surface by bone marrow-derived human mesenchymal stem cells. Stem Cells 2006, 24, 315–321. [Google Scholar] [CrossRef] [PubMed]
  50. Jiang, T.S.; Cai, L.; Ji, W.Y.; Hui, Y.N.; Wang, Y.S.; Hu, D.; Zhu, J. Reconstruction of the corneal epithelium with induced marrow mesenchymal stem cells in rats. Mol. Vis. 2010, 16, 1304–1316. [Google Scholar] [PubMed]
  51. Gu, S.; Xing, C.; Han, J.; Tso, M.O.; Hong, J. Differentiation of rabbit bone marrow mesenchymal stem cells into corneal epithelial cells in vivo and ex vivo. Mol. Vis. 2009, 15, 99–107. [Google Scholar] [PubMed]
  52. Omoto, M.; Miyashita, H.; Shimmura, S.; Higa, K.; Kawakita, T.; Yoshida, S.; McGrogan, M.; Shimazaki, J.; Tsubota, K. The use of human mesenchymal stem cell-derived feeder cells for the cultivation of transplantable epithelial sheets. Invest. Ophthalmol. Vis. Sci. 2009, 50, 2109–2115. [Google Scholar] [CrossRef] [PubMed]
  53. Hu, N.; Zhang, Y.Y.; Gu, H.W.; Guan, H.J. Effects of bone marrow mesenchymal stem cells on cell proliferation and growth factor expression of limbal epithelial cells in vitro. Ophthalmic Res. 2012, 48, 82–88. [Google Scholar] [CrossRef] [PubMed]
  54. Martínez-Conesa, E.M.; Espel, E.; Reina, M.; Casaroli-Marano, R.P. Characterization of ocular surface epithelial and progenitor cell markers in human adipose stromal cells derived from lipoaspirates. Invest. Ophthalmol. Vis. Sci. 2012, 53, 513–520. [Google Scholar] [CrossRef] [PubMed]
  55. Nieto-Miguel, T.; Galindo, S.; Reinoso, R.; Corell, A.; Martino, M.; Pérez-Simón, J.A.; Calonge, M. In vitro simulation of corneal epithelium microenvironment induces a corneal epithelial-like cell phenotype from human adipose tissue mesenchymal stem cells. Curr. Eye Res. 2013, 38, 933–944. [Google Scholar] [CrossRef] [PubMed]
  56. Ho, J.H.; Ma, W.H.; Tseng, T.C.; Chen, Y.F.; Chen, M.H.; Lee, O.K. Isolation and characterization of multi-potent stem cells from human orbital fat tissues. Tissue Eng. Part A 2011, 17, 255–266. [Google Scholar] [CrossRef] [PubMed]
  57. Lin, K.J.; Loi, M.X.; Lien, G.S.; Cheng, C.F.; Pao, H.Y.; Chang, Y.C.; Ji, A.T.; Ho, J.H. Topical administration of orbital fat-derived stem cells promotes corneal tissue regeneration. Stem Cell Res. Ther. 2013, 4. [Google Scholar] [CrossRef] [PubMed]
  58. Chen, S.Y.; Mahabole, M.; Horesh, E.; Wester, S.; Goldberg, J.L.; Tseng, S.C. Isolation and Characterization of Mesenchymal Progenitor Cells from Human Orbital Adipose Tissue. Invest Ophthalmol. Vis. Sci. 2014, 55, 4842–4852. [Google Scholar] [CrossRef] [PubMed]
  59. Monteiro, B.G.; Serafim, R.C.; Melo, G.B.; Silva, M.C.; Lizier, N.F.; Maranduba, C.M.; Smith, R.L.; Kerkis, A.; Cerruti, H.; Gomes, J.A.; et al. Human immature dental pulp stem cells share key characteristic features with limbal stem cells. Cell Prolif. 2009, 42, 587–594. [Google Scholar] [CrossRef] [PubMed]
  60. Gomes, J.A.; Geraldes Monteiro, B.; Melo, G.B.; Smith, R.L.; Cavenaghi Pereira da Silva, M.; Lizier, N.F.; Kerkis, A.; Cerruti, H.; Kerkis, I. Corneal reconstruction with tissue-engineered cell sheets composed of human immature dental pulp stem cells. Invest. Ophthalmol. Vis. Sci. 2010, 51, 1408–1414. [Google Scholar] [CrossRef] [PubMed]
  61. Blazejewska, E.A.; Schlotzer-Schrehardt, U.; Zenkel, M.; Bachmann, B.; Chankiewitz, E.; Jacobi, C.; Kruse, F.E. Corneal limbal microenvironment can induce transdifferentiation of hair follicle stem cells into corneal epithelial-like cells. Stem Cells 2009, 27, 642–652. [Google Scholar] [CrossRef] [PubMed]
  62. Meyer-Blazejewska, E.A.; Call, M.K.; Yamanaka, O.; Liu, H.; Schlotzer-Schrehardt, U.; Kruse, F.E.; Kao, W.W. From hair to cornea: Toward the therapeutic use of hair follicle-derived stem cells in the treatment of limbal stem cell deficiency. Stem Cells 2011, 29, 57–66. [Google Scholar] [CrossRef] [PubMed]
  63. Reza, H.M.; Ng, B.Y.; Phan, T.T.; Tan, D.T.; Beuerman, R.W.; Ang, L.P. Characterization of a novel umbilical cord lining cell with CD227 positivity and unique pattern of P63 expression and function. Stem Cell Rev. 2011, 7, 624–638. [Google Scholar] [CrossRef] [PubMed]
  64. Reza, H.M.; Ng, B.Y.; Gimeno, F.L.; Phan, T.T.; Ang, L.P. Umbilical cord lining stem cells as a novel and promising source for ocular surface regeneration. Stem Cell Rev. 2011, 7, 935–947. [Google Scholar] [CrossRef] [PubMed]
  65. Kanayama, S.; Nishida, K.; Yamato, M.; Hayashi, R.; Sugiyama, H.; Soma, T.; Maeda, N.; Okano, T.; Tano, Y. Analysis of angiogenesis induced by cultured corneal and oral mucosal epithelial cell sheets in vitro. Exp. Eye Res. 2007, 85, 772–781. [Google Scholar] [CrossRef] [PubMed]
  66. Kanayama, S.; Nishida, K.; Yamato, M.; Hayashi, R.; Maeda, N.; Okano, T.; Tano, Y. Analysis of soluble vascular endothelial growth factor receptor-1 secreted from cultured corneal and oral mucosal epithelial cell sheets in vitro. Br. J. Ophthalmol. 2009, 93, 263–267. [Google Scholar] [CrossRef] [PubMed]
  67. ClinicalTrials.gov, a service of US National Institute of Health. Available online: https://clinicaltrials.gov/ (accessed 30 December 2014).
  68. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Yu, J.; Vodyanik, M.A.; Smuga-Otto, K.; Antosiewicz-Bourget, J.; Frane, J.L.; Tian, S.; Nie, J.; Jonsdottir, G.A.; Ruotti, V.; Stewart, R.; et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007, 318, 1917–1920. [Google Scholar] [CrossRef] [PubMed]
  70. Yamanaka, S. Induced pluripotent stem cells: Past, present, and future. Cell Stem Cell 2012, 10, 678–684. [Google Scholar] [CrossRef] [PubMed]
  71. Kelaini, S.; Cochrane, A.; Margariti, A. Direct reprogramming of adult cells: Avoiding the pluripotent state. Stem Cells Cloning 2014, 15, 19–29. [Google Scholar]
  72. Pietronave, S.; Prat, M. Advances and applications of induced pluripotent stem cells. Can. J. Physiol. Pharmacol. 2012, 90, 317–325. [Google Scholar] [CrossRef] [PubMed]
  73. Yang, K.; Jiang, Z.; Wang, D.; Lian, X.; Yang, T. Corneal epithelial-like transdifferentiation of hair follicle stem cells is mediated by pax6 and beta-catenin/Lef-1. Cell Biol. Int. 2009, 33, 861–866. [Google Scholar] [CrossRef] [PubMed]
  74. Saichanma, S.; Bunyaratvej, A.; Sila-Asna, M. In vitro transdifferentiation of corneal epithelial-like cells from human skin-derived precursor cells. Int. J. Ophthalmol. 2012, 5, 158–163. [Google Scholar] [PubMed]
  75. Casaroli-Marano, R.P.; Zarbin, M.A. Cell-based therapy for retinal degenerative disease. In Developments in Ophthalmology; Karger: Basel, Switzerland, 2014; Volume 53. [Google Scholar]
  76. Zhong, X.; Gutierrez, C.; Xue, T.; Hampton, C.; Vergara, M.N.; Cao, L.H.; Peters, A.; Park, T.S.; Zambidis, E.T.; Meyer, J.S.; et al. Generation of three-dimensional retinal tissue with functional photoreceptors from human iPSCs. Nat. Commun. 2014, 10. [Google Scholar] [CrossRef] [Green Version]
  77. Mekala, S.R.; Vauhini, V.; Nagarajan, U.; Maddileti, S.; Gaddipati, S.; Mariappan, I. Derivation, characterization and retinal differentiation of induced pluripotent stem cells. J. Biosci. 2013, 38, 123–134. [Google Scholar] [CrossRef] [PubMed]
  78. Notara, M.; Hernandez, D.; Mason, C.; Daniels, J.T. Characterization of the phenotype and functionality of corneal epithelial cells derived from mouse embryonic stem cells. Regen. Med. 2012, 7, 167–178. [Google Scholar] [CrossRef] [PubMed]
  79. Yu, D.; Chen, M.; Sun, X.; Ge, J. Differentiation of mouse induced pluripotent stem cells into corneal epithelial-like cells. Cell Biol. Int. 2013, 37, 87–94. [Google Scholar] [CrossRef] [PubMed]
  80. Hayashi, R.; Ishikawa, Y.; Ito, M.; Kageyama, T.; Takashiba, K.; Fujioka, T.; Tsujikawa, M.; Miyoshi, H.; Yamato, M.; Nakamura, Y.; et al. Generation of corneal epithelial cells from induced pluripotent stem cells derived from human dermal fibroblast and corneal limbal epithelium. PLoS One 2012, 7, e45435. [Google Scholar] [CrossRef] [PubMed]
  81. Sareen, D.; Saghizadeh, M.; Ornelas, L.; Winkler, M.A.; Narwani, K.; Sahabian, A.; Funari, V.A.; Tang, J.; Spurka, L.; Punj, V.; et al. Differentiation of Human Limbal-Derived Induced Pluripotent Stem Cells Into Limbal-Like Epithelium. Stem Cells Transl. Med. 2014, 3, 1002–1012. [Google Scholar] [CrossRef] [PubMed]
  82. Mikhailova, A.; Ilmarinen, T.; Uusitalo, H.; Skottman, H. Small-molecule induction promotes corneal epithelial cell differentiation from human induced pluripotent stem cells. Stem Cell Rep. 2014, 6, 219–231. [Google Scholar] [CrossRef]
  83. Zhu, J.; Zhang, K.; Sun, Y.; Gao, X.; Li, Y.; Chen, Z.; Wu, X. Reconstruction of functional ocular surface by acellular porcine cornea matrix scaffold and limbal stem cells derived from human embryonic stem cells. Tissue Eng. Part A 2013, 19, 2412–2425. [Google Scholar] [CrossRef] [PubMed]
  84. Taylor, C.J.; Peacock, S.; Chaudhry, A.N.; Bradley, J.A.; Bolton, E.M. Generating an iPSC bank for HLA-matched tissue transplantation based on known donor and recipient HLA types. Cell Stem Cell 2012, 11, 147–152. [Google Scholar] [CrossRef] [PubMed]
  85. Alexandrova, E.M.; Petrenko, O.; Nemajerova, A.; Romano, R.A.; Sinha, S.; Moll, U.M. ΔNp63 regulates select routes of reprogramming via multiple mechanisms. Cell Death Differ. 2013, 20, 1698–1708. [Google Scholar] [CrossRef] [PubMed]
  86. Ng, W.L.; Chen, G.; Wang, M.; Wang, H.; Story, M.; Shay, J.W.; Zhang, X.; Wang, J.; Amin, A.R.; Hu, B.; et al. OCT4 as a target of miR-34a stimulates p63 but inhibits p53 to promote human cell transformation. Cell Death Dis. 2014, 5. [Google Scholar] [CrossRef]
  87. Shalom-Feuerstein, R.; Serror, L.; Aberdam, E.; Müller, F.J.; van Bokhoven, H.; Wiman, K.G.; Zhou, H.; Aberdam, D.; Petit, I. Impaired epithelial differentiation of induced pluripotent stem cells from ectodermal dysplasia-related patients is rescued by the small compound APR-246/PRIMA-1MET. Proc. Natl. Acad. Sci. USA 2013, 110, 2152–2156. [Google Scholar] [CrossRef] [PubMed]
  88. Shalom-Feuerstein, R.; Serror, L.; de La Forest Divonne, S.; Petit, I.; Aberdam, E.; Camargo, L.; Damour, O.; Vigouroux, C.; Solomon, A.; Gaggioli, C.; et al. Pluripotent stem cell model reveals essential roles for miR-450b-5p and miR-184 in embryonic corneal lineage specification. Stem Cell 2012, 30, 898–909. [Google Scholar] [CrossRef]
  89. Li, X.; Pei, D.; Zheng, H. Transitions between epithelial and mesenchymal states during cell fate conversions. Protein Cell 2014, 5, 580–591. [Google Scholar] [CrossRef] [PubMed]
  90. Grocott, T.; Johnson, S.; Bailey, A.P.; Streit, A. Neural crest cells organize the eye via TGF-β and canonical Wnt signalling. Nat. Commun. 2011, 2. [Google Scholar] [CrossRef]
  91. Chan, A.A.; Hertsenberg, A.J.; Funderburgh, M.L.; Mann, M.M.; Du, Y.; Davoli, K.A.; Mich-Basso, J.D.; Yang, L.; Funderburgh, J.L. Differentiation of human embryonic stem cells into cells with corneal keratocyte phenotype. PLoS One 2013, 8, e56831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Chien, Y.; Liao, Y.W.; Liu, D.M.; Lin, H.L.; Chen, S.J.; Chen, H.L.; Peng, C.H.; Liang, C.M.; Mou, C.Y.; Chiou, S.H. Corneal repair by human corneal keratocyte-reprogrammed iPSCs and amphiphatic carboxymethyl-hexanoyl chitosan hydrogel. Biomaterials 2012, 33, 8003–8016. [Google Scholar] [CrossRef] [PubMed]
  93. Fukuta, M.; Nakai, Y.; Kirino, K.; Nakagawa, M.; Sekiguchi, K.; Nagata, S.; Matsumoto, Y.; Yamamoto, T.; Umeda, K.; Heike, T.; et al. Derivation of Mesenchymal Stromal Cells from Pluripotent Stem Cells through a Neural Crest Lineage using Small Molecule Compounds with Defined Media. PLoS One 2014, 9, e112291. [Google Scholar] [CrossRef] [PubMed]
  94. Gore, A.; Li, Z.; Fung, H.; Young, J.E.; Agarwal, S.; Antosiewicz-Bourget, J.; Canto, I.; Giorgetti, A.; Israel, M.A.; Kiskinis, E.; et al. Somatic coding mutations in human induced pluripotent stem cells. Nature 2011, 471, 63–67. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Casaroli-Marano, R.P.; Nieto-Nicolau, N.; Martínez-Conesa, E.M.; Edel, M.; B.Álvarez-Palomo, A. Potential Role of Induced Pluripotent Stem Cells (IPSCs) for Cell-Based Therapy of the Ocular Surface. J. Clin. Med. 2015, 4, 318-342. https://doi.org/10.3390/jcm4020318

AMA Style

Casaroli-Marano RP, Nieto-Nicolau N, Martínez-Conesa EM, Edel M, B.Álvarez-Palomo A. Potential Role of Induced Pluripotent Stem Cells (IPSCs) for Cell-Based Therapy of the Ocular Surface. Journal of Clinical Medicine. 2015; 4(2):318-342. https://doi.org/10.3390/jcm4020318

Chicago/Turabian Style

Casaroli-Marano, Ricardo P., Núria Nieto-Nicolau, Eva M. Martínez-Conesa, Michael Edel, and Ana B.Álvarez-Palomo. 2015. "Potential Role of Induced Pluripotent Stem Cells (IPSCs) for Cell-Based Therapy of the Ocular Surface" Journal of Clinical Medicine 4, no. 2: 318-342. https://doi.org/10.3390/jcm4020318

Article Metrics

Back to TopTop