Next Article in Journal
Preparation of Defined Chitosan Oligosaccharides Using Chitin Deacetylases
Next Article in Special Issue
JAK/STAT Activation: A General Mechanism for Bone Development, Homeostasis, and Regeneration
Previous Article in Journal
Molecular Mechanisms of SGLT2 Inhibitor on Cardiorenal Protection
Previous Article in Special Issue
Comparative Proteomic Analysis Identifies EphA2 as a Specific Cell Surface Marker for Wharton’s Jelly-Derived Mesenchymal Stem Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Differentiation Induction of Human Stem Cells for Corneal Epithelial Regeneration

by
Kasem Theerakittayakorn
1,†,
Hong Thi Nguyen
1,†,
Jidapa Musika
1,
Hataiwan Kunkanjanawan
2,
Sumeth Imsoonthornruksa
1,
Sirilak Somredngan
1,
Mariena Ketudat-Cairns
1 and
Rangsun Parnpai
1,*
1
Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
2
Medeze Research and Development Co., Ltd. 28/9 Moo 8, Phutthamonthon Sai 4 Rd., Krathum Lom, Sam Phran, Nakhon Pathom 73220, Thailand
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2020, 21(21), 7834; https://doi.org/10.3390/ijms21217834
Submission received: 25 September 2020 / Revised: 16 October 2020 / Accepted: 17 October 2020 / Published: 22 October 2020

Abstract

:
Deficiency of corneal epithelium causes vision impairment or blindness in severe cases. Transplantation of corneal epithelial cells is an effective treatment but the availability of the tissue source for those cells is inadequate. Stem cells can be induced to differentiate to corneal epithelial cells and used in the treatment. Multipotent stem cells (mesenchymal stem cells) and pluripotent stem cells (embryonic stem cells and induced pluripotent stem cells) are promising cells to address the problem. Various protocols have been developed to induce differentiation of the stem cells into corneal epithelial cells. The feasibility and efficacy of both human stem cells and animal stem cells have been investigated for corneal epithelium regeneration. However, some physiological aspects of animal stem cells are different from those of human stem cells, the protocols suited for animal stem cells might not be suitable for human stem cells. Therefore, in this review, only the investigations of corneal epithelial differentiation of human stem cells are taken into account. The available protocols for inducing the differentiation of human stem cells into corneal epithelial cells are gathered and compared. Also, the pathways involving in the differentiation are provided to elucidate the relevant mechanisms.

1. Introduction

Cornea is the transparent anterior part of the eye. Its transparency allows light to enter into the eye and the visual sensation will be initiated once the light touches the photoreceptor cells in the retina. The curvature of the cornea plays an important role in light refraction. It has a refractive power of approximately 60 diopters or two-thirds of the total eye refractive power [1]. The cornea is the first part of the eye that touches the light and it is exposed to the external environment, therefore it is easily damaged more than other parts of the eye. It can be injured by chemicals, heat, and forces from accidents. The outer surface of the cornea is covered by epithelial tissue in which the corneal epithelial cells are generated from the stem cells at the corneoscleral junction called the limbus. Limbal stem cells, the stem cells residing in the limbus, generate new corneal epithelial cells to replace the shed old cells or damaged cells to maintain the corneal epithelial population. Severe damage, severe microbial infections, or some diseases such as Stevens–Johnson syndrome and ocular cicatricial pemphigoid [2,3] may cause the depletion of limbal stem cells. Such a condition is called limbal stem cell deficiency (LSCD). Once the limbal stem cells are lost, corneal epithelial cells are unable to be generated and it will result in visual impairment and finally vision loss.
Many medical treatments have been investigated to restore the corneal epithelium. The common strategy is the transplantation of autologous epithelial cells from the contralateral cornea [4,5]. In this strategy, limbal tissue is biopsied from the healthy cornea. Then, epithelial cells are isolated and expanded in laboratory until enough cells are obtained for transplantation. Even though this procedure is effective and has high success rate of 70–80% [4], it is unable to be applied to patients with bilateral LSCD. Transplantation of allogeneic limbal epithelial cells is an optional strategy but it has a risk of graft rejection [6,7,8]. To avoid the lack of autologous limbal epithelium and high risk of graft failure from allogeneic sources, autologous stem cells become a promising option for the regeneration of corneal epithelial tissue.
With the differentiation capacity, stem cells can be induced to be corneal epithelial cells and used in medical treatments. Various kinds of stem cells have been studied for corneal epithelial tissue regeneration [9,10,11]. Mesenchymal stem cells (MSCs), which possess multipotent differentiation capacity, can be isolated from patients and used in the treatments without the concern of immune rejection [12]. By using minimally invasive procedures and uncomplicated techniques, MSCs can be obtained from diverse tissues, including bone marrow, adipose tissue, dental pulp, umbilical cord, and amniotic fluid [13,14,15]. However, it is difficult to induce MSCs from mesoderm to undergo transdifferentiation into corneal epithelial cells which originally derived from ectoderm. Pluripotent stem cells (PSCs) such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) are also of interest. They are unlimited self-renewal and have the potential to differentiate into any cell type of the adult organs [16,17]. However, complicated techniques and expensive materials are required for obtaining PSCs [18,19].
Several publications reported about the capability of MSCs and PSCs to differentiate into corneal epithelial cells [20,21,22,23,24,25]. However, the methods used in the differentiation induction are very diverse. Until now, no standard differentiation protocol is available and most published researches studied stem cells from animals. Because animal stem cells are different from human stem cells in many physiological aspects [26,27,28], the protocols developed from the studies of animal stem cells may not be suitable for human stem cells [29,30]. Therefore, herein, we review the progress of the corneal epithelial differentiation of only human stem cells. We also provide fundamental knowledge related to the differentiation methods which include the nature of corneal epithelial cells and the relevant differentiation pathways.

2. Corneal Epithelial Cell

The cornea consists of three cellular layers: epithelium, stroma, and endothelium [31]. Bowman’s membrane is the thin matrix layer between epithelium and stroma whereas Descemet’s membrane is the matrix layer between stroma and endothelium [32]. The corneal epithelium covers the outermost surface of the cornea and protects the cornea from pathogen invasion [33]. The corneal epithelium is derived from surface ectoderm at 5–6 weeks of gestation [34]. There are 5–6 layers of non-keratinized stratified epithelial cells [35,36] in the corneal epithelial tissue which has a thickness of approximately 50 µm [37]. The corneal epithelium consists of three types of epithelial cells: basal cells, wing cells, and superficial cells [38]. The basal cells are arranged in a single cell layer adhered to the underlying Bowman’s membrane. They have cuboidal or columnar shape and have the ability for mitotic cell division [34]. Wing cells are in the suprabasal layers covering the layer of basal cells. They are polyhedral cells stratified 1–3 layers [39]. The superficial cells cover the outer surface of cornea. They are squamous cells with flattened nuclei. Terminal differentiation of the superficial cells results in cells sloughing off into the tears [40,41].
The integrity of corneal epithelium requires the balance between cellular proliferation and differentiation. While corneal epithelial cells are continuously desquamated from the corneal surface into the tear film, the new corneal epithelial cells are continually generated from the basal cells [37,42] and the stem cells residing at the limbus, the transitional area between the cornea and the sclera [43]. The regeneration of new corneal epithelial cells is achieved by the proliferation, migration, and differentiation of the corneal stem cells. The stem cells will divide into transient amplifying cells. The transient amplifying cells, which have rapid mitotic activity, will proliferate to get enough cell number before terminal differentiation [44]. The newly generated cells from the basal cells migrate vertically to the corneal surface and those from limbal stem cells migrate centripetally to the central cornea [45]. The differentiation process requires 7–10 days before they undergo apoptosis and desquamation [34]. By the delicate control, the rate of cell proliferation equals the rate of cell desquamation. Consequently, the mass of the corneal epithelium remains constant.
The epithelial cells of cornea, limbus, and conjunctiva possess distinct characteristics. Cytokeratins (CKs), which are intermediate filaments expressed in a tissue-specific manner [46], are usually used to distinguish corneal epithelial cells from other cell types. There are about 20 types of CKs in humans [47]. The expressions of CK1–CK10 and CK13–CK20 of the corneal, limbal, and conjunctival epithelial cells were clarified by Reinstein Merjava and colleagues [37]. One cytokeratin type can be expressed in more than one cell types but CK3 and CK12 are expressed only in corneal epithelial cells and commonly used as specific markers for corneal epithelial cells [35,47,48,49,50]. Besides CKs, corneal epithelial cells at each stage of differentiation can be characterized by some specific markers. Involucrin is expressed in the differentiated superficial corneal epithelial cells. The expression of involucrin is low in the limbus but high in the central cornea [51,52]. Epidermal growth factor receptor (EGFR) is expressed at the basal layer of corneal epithelium while transglutaminase 1 (TG1) is expressed in the upper layer [53]. Connexin 43 (Cx43), a gap junction protein which is acquired during corneal epithelial differentiation, is expressed in the basal layer of the corneal epithelium but it is not expressed in the limbus [35]. Limbal epithelial cells express CK5, CK14 [54], and some putative stem cell markers such as tumor protein (p63), ATP-binding cassette transporter G2 (ABCG2), and CK19. These stem cells markers of limbal epithelial cells will gradually decrease and finally disappear when they become transient amplifying cells and migrate to the central cornea [55].

3. Signaling Pathways Related to Corneal Epithelial Differentiation

The connection of the activities of Wnt/β-catenin, transforming growth factor beta (TGF-β)/Nodal, and bone morphogenetic protein (BMP) signaling pathway is necessary for the formation of the embryonic murine head [56]. Moreover, the cross-linkage of retinoic acid (RA) signaling, Wnt/β-catenin, paired-like homeodomain 2 (Pitx2) is essential for early eye development [57].
Wnt signaling plays an important role in eye development and its signaling is variable between the different periods [58]. Inhibiting or blocking Wnt signaling results in differentiation into epithelial cells [59,60,61]. Suppression of Wnt signaling can be done via inhibiting the initiation of Wnt signaling, stabilization, and translocation of β-catenin into cell nucleus (Figure 1). Initiation of Wnt signaling is downregulated by blocking low-density lipoprotein-related receptors 5 and 6 (LRP5/6 receptor) via Dickkopt (Dkk) proteins (Dkk1 [62,63], Dkk2 [64], Dkk3 [65]) or Frizzled 7 receptor by ring finger protein 43 (RNF43) [66], secreted Frizzled-related proteins (SFRP) [63]) or reducing Wnt secretion [67]. Reducing expression of Dkk2 via generating Dkk2-null mice increase the Wnt signaling target gene (axis inhibition protein 2 (Axin2)), leading to a decrease of corneal differentiation in ocular surface ectoderm of mouse embryo [64]. Corneal specific marker (CK12) expression is low and not continuous in the corneal epithelium of Dkk2 mutant mouse while its expression is high and continuous in wide type mouse [64]. Paired box protein (PAX6), the key transcription factor regulated eye development and maintenance of the adult cornea [41], can prevent activation of Wnt/β-catenin [58,63,68] via increasing expression of few Wnt inhibitors (Dkk1, SFRP1, SFRP2 [63], and Dkk3 [65]). Overexpression of PAX6 via its transfection into ESCs or adult stem cells induced corneal epithelium-like cells [69,70]. Two PAX6 isoforms differently induce expression of corneal specific genes (CK3 and CK12) [71]. While PAX6 isoform-a combined with Klf4 induces CK3 expression, its isoform-b combined with Kruppel-like factor 4 (Klf4) and octamer-binding transcription factor 4 (Oct4) induces CK12 expression [71]. Especially, two PAX6 isoforms combined with Klf4 and Oct4 cooperatively induce both CK3 and CK12 although induction efficiency relies on cell types [71]. Other eye field transcription factors (retinal homeobox protein (Rax), SIX homeobox 3 (Six3), homeodomain protein Optx2) also be mention that they suppress activation of Wnt signaling [58,72] but their effects on corneal differentiation have not yet reported. Another way for downregulating the initiation of Wnt signaling is inhibition of Wnt secretion by using inhibitors of porcupine, an essential enzyme for post-translational modification of Wnt, such as ETC-159, C59, IWPs, LGK974 [67]. By preventing the activity of porcupine, IWPs (IWP1, 2, 3 and 4) reduce β-catenin content in the cytoplasm [73]. Supplementation of IWP2 in low calcium medium can differentiate limbal epithelial progenitor isolated from enzymatic digestion of human limbal biopsy fragments into corneal epithelial cells expressed CK12 mRNA and protein [74]. IWP2 downregulates β-catenin content and further reduces expression of stem/progenitor markers (p63 and ABCG2). However, the effect of IWP2 on corneal epithelium differentiation depends on the culture medium and the isolated method. IWP2 increases p63 and ABCG2 expression and colony formation in the limbal epithelial progenitor cells isolated from limbal explants cultured in the supplemented hormonal epithelial medium [74].
When Wnt cannot bind with dual receptors (LRP5/6 and Frizzled 7), the level of β-catenin will be reduced by the β-catenin destruction complex which phosphorylates β-catenin, leading to ubiquitination and further degradation of β-catenin [73,75]. The β-catenin destruction complex contains glycogen synthase kinase-3 (GSK3), casein kinase 1, adenomatous polyposis coli (APC), and Axin [75]. IWR compounds (IWR 1, 2, 3, 4, 5) decrease β-catenin by upregulating β-catenin degradation via increasing the stability of Axin2 protein that is included in the β-catenin destruction complex [73]. IWR and other small molecules (XAV939, G007-LK) promote stability of Axin via inhibition Tankyrase enzyme which degrades Axin [67]. Although using of IWR1 in combination with other factors induces differentiation into corneal epithelial cells [76], using IWR1 alone does not affect lens regeneration [77].
Translocation of β-catenin is inhibited by small molecules such as RA, SU666 (the Src family kinase inhibitor), leading to a decrease in the level of β-catenin in the cell nucleus and Wnt target genes [59]. Both small molecules can drive human PSCs into simple epithelial cells that expressed CK18, CK8 [59].
The Wnt signaling pathway is blocked by deleting β-catenin or LRP5/6 receptor. Knockout β-catenin or LRP5/6 receptor can induce corneal epithelium from corneal stroma keratinocytes [60]. Knockout β-catenin or LRP5/6 receptor increase the expression of BMP4 that further enhances expression of p63, phosphorylated Smad1/5 (pSmad1/5), and phosphorylated extracellular regulated kinase 1/2 (pERK1/2), leading to corneal epithelial stratification [60].
TGF-β family members (TGF-βs, activin, and BMPs) also regulate epithelial differentiation in eye development [78]. Suppression TGF-β signaling and/or activating BMP signaling, together with another signaling (Wnt signaling or epidermal growth factor (EGF) signaling or fibroblast growth factor (FGF) signaling), are crucial for generating corneal epithelial cells from PSCs. SB505124, SB431542, A83-01, A77-01 are selective inhibitors of activin and TGF-β signaling pathway. These small molecules inhibit cytoplasmic signal transducers (Smad2 and Smad3) via inhibition of TGF-β type I receptors (activin receptor-like kinases (ALKs): ALK4, ALK5, and ALK7) [79,80,81] but SB505124 and SB431542 do not inhibit other receptors (ALK1, ALK2, ALK3, and ALK6) involved BMP signaling [79,80] while A83-01 has a slight effect on BMP receptor [81]. SB431542 supplementation improved the proliferation and maintenance phenotype of mouse lens epithelial cells [82]. Moreover, SB431542 treatment promotes the BMP activity of non-responsive human induced pluripotent stem cell line [83]. BMP signaling plays an important role in lens epithelial differentiation [78]. BMP4 or BMP7 treatment promotes the phosphorylation of Smad1/5/8 and further enhance BMP signaling [84]. Blocking TGF-β signaling and enhancing BMP and FGF signaling induce corneal limbal stem cells [85,86,87]. Treatment with blebbistatin (a non-myosin II inhibitor) at day 1, SB505124 (an inhibitor of TGF-β signaling) and basic FGF (bFGF, an activator of FGF signaling) at day 2, and BMP4 (an activator of BMP signaling) at day 3–4 drive human PSCs into surface ectodermal cells that will further differentiate into corneal limbal stem cells expressed p63, CK15 [85], ABCG2, Leucin-rich repeat-containing G-protein couple receptor 5 (LGR5), CD200 [87]. Moreover, these cells express CK3 (a specific corneal marker) after stratification culture [85]. Other co-operation of signaling pathways, blocking TGF-β and Wnt signaling and promoting FGF signaling, generates corneal epithelial cells from human iPSCs [61,76]. Supplementation of SB505124, IWP2, and bFGF in the induction medium produces high differentiation efficiency of hiPSCs into corneal epithelial cells that were positively stained with both specific corneal markers (CK12 and CK3) [61]. Treatment human ESCs with IWR1 (an inhibitor of Wnt signaling), A83-01, and bFGF can generate corneal epithelial cells expressed CK12 and CK3 [76] but the differentiated time is longer than Mikhailova et al. (2014) that may be caused by A83-01 slightly inhibited BMP signaling. Blocking Wnt signaling and activating BMP and FGF signaling by using RA, BMP4, and bFGF can generate corneal epithelial cells expressed CK3 [59]. Blocking Wnt signaling and activating BMP and EGF signaling via treatment human PSCs with RA, BMP4, and EGF can induce corneal epithelial-like cells [83]. A summary of signaling pathways involved in generation corneal epithelial cells is shown in Figure 1.

4. Corneal Epithelial Differentiation of Human MSCs

MSCs are widely used for cell therapies and regenerative medicine because of their great advantages of multipotent differentiation capacity, easy accessibility, and easy manipulation [88,89,90,91,92,93]. MSCs can be obtained from various adult tissues (e.g., bone marrow, adipose tissue, dental pulp, periodontal ligament, dermis, muscle) and fetal tissues (e.g., umbilical cord, placenta, amniotic fluid). According to the minimal criteria for defining MSCs by International Society for Cell & Gene Therapy (ISCT), MSCs are plastic-adherent cells with the capability of in vitro differentiation into adipocytes, chondrocytes, and osteoblasts. Moreover, MSCs must express CD73, CD90 and CD105 but not express CD11b, CD14, CD19, CD34, CD45, CD79a and HLA-DR [94]. Even though MSCs originate from mesoderm [95], MSCs are also able to differentiate into the cells of both endodermal and ectodermal lineages [96,97,98,99]. Likewise, MSCs can differentiate in vitro into corneal epithelial cells which are the cells in ectodermal linage.
To treat LSCD, transplantation of corneal or limbal epithelial cells are the common and effective procedures [100]. But the corneal tissues for isolating those cells are limited, thus there are attempts to use MSCs instead. Transplantations of MSCs for LSCD treatment in animal models have been being investigated and the results revealed satisfying outcomes. The investigated human MSCs were derived from various sources. Human bone marrow-derived MSCs (BM-MSCs) were grown on the amniotic membrane and transplanted in rats [7] or rabbits [101]. Human MSCs from immature dental pulps were prepared as cell sheets and transplanted in rabbits [102,103]. Human adipose tissue-derived MSCs (AT-MSCs) from orbital fat were directly transplanted by topical administration in mice [104]. Also, human AT-MSCs were directly transplanted by topical administration in rats [105] or grown on the amniotic membrane and transplanted in rabbits [106]. After transplantation, the epithelia were regenerated, the ocular surfaces were recovered, and corneal transparency was improved. The positive expression of CK3 or CK12 confirmed the terminal differentiation of the transplanted MSCs into corneal epithelial cells. Even though MSCs were transplanted without prior differentiation induction, the differentiation can be initiated because the cells at the transplantation sites can secrete signaling molecules directing them to become corneal epithelial cells [107,108,109]. Unfortunately, in some experiments, terminal differentiation was very low [104] or cannot be obtained [7]. Probably, the degree of differentiation after transplantation depends on the biological microenvironment at the transplantation sites which are difficult to be controlled [110,111,112].
In vitro differentiation induction can be achieved by using undefined induction medium and defined induction medium [113,114]. The undefined induction medium can be obtained from signaling molecules secreted by some appropriate cell types. The cells from cornea, both corneal cells and limbal cells, can provide signaling molecules regulating corneal epithelial differentiation [115,116,117]. Therefore, they are used to drive MSCs to differentiate into corneal epithelial cells in laboratories. In order to use signaling molecules from those cells, two methods are applicable; conditioned medium (CM) and co-culture system. In the first way, the corneal epithelial cells or limbal cells grown in culture containers will release inducing substances into the culture medium which can be harvested and used as CM. Once MSCs are cultured in the CM, MSCs will be induced to differentiate into corneal epithelial cells [118]. Another way is the co-culture system in which MSCs and the signal providing cells are cultured together in cell culture insert system. In such a culture system, one cell type is grown on the bottom surface of the culture container whereas another cell type is grown on the membrane surface of an equipment inserted in the same culture container [24,119,120]. The signaling molecules from one cell type can diffuse through the membrane pore to another cell type. In both strategies, the components in the medium cannot be defined and the signaling molecules playing roles in the differentiation induction cannot be specified.
Using CM as undefined induction medium was investigated in human AT-MSCs [118]. In this experiment, they compared the effects of the CM from human corneal epithelial cells and CM from human limbal fibroblasts on the corneal epithelial differentiation of AT-MSCs. Unfortunately, they found that AT-MSCs initially expressed CK3 and CK12. After AT-MSCs were cultured in the CMs, no significant difference was found in the expression of CK3. However, the expression of CK12 significantly increased after AT-MSCs were cultured in the CM from human corneal epithelial cells.
The co-culture system was applied in the differentiation of AT-MSCs [119], conjunctiva-derived MSCs (CJ-MSCs) [24], and exfoliated deciduous teeth-derived MSCs (EDT-MSCs) [120]. The MSCs in the experiments can undergo terminal corneal epithelial differentiation as shown by the results of gene expression or immunocytochemistry (ICC) of CK3 and CK12 markers. The details of the co-culture experiments are concluded in Table 1.
Using undefined induction medium from the aforementioned strategies need to culture signal providing cells in which there are some disadvantages; complex manipulation, expensive equipment in the culture insert system, and uncontrollable medium compositions. By contrast, defined induction media consist of known ingredient proportion and their ingredients can be adjusted to be suitable for specific cells. Moreover, defined induction media also can be used to investigate the effects of individual inducing molecules on the corneal epithelial differentiation. Various growth factors and bioactive molecules were added as ingredients of defined induction media. The defined induction media, which were used in the differentiation of human MSCs into corneal epithelial cells, and their results are summarized in Table 2.
In the differentiation of human CJ-MSCs by Soleimanifar et al. [24], they compared the differentiation outcome obtained from the co-culture as shown in Table 1 with the outcome obtained from the induction medium as shown in Table 2. According to their induction conditions, the co-culture offered much better differentiation outcome than the induction medium. However, the better outcome from co-culture possibly came from the effect of some bioactive molecules in the culture medium of the co-culture experiment. Because, when CnT-Prime medium was used in co-culture, the differentiation outcome from co-culture was poorer than the outcome from the induction medium. In the differentiation of human bone marrow-derived MSCs (BM-MSCs) by Katikireddy et al. [20], they determined the differentiation capacity between sorted SSEA4+ BM-MSCs and unsorted BM-MSCs in two-step induction in which different induction medium was used in each step. They found that sorted SSEA4+ BM-MSCs underwent corneal epithelial differentiation more than unsorted BM-MSCs. Nieto-Nicolau et al. [21] induced corneal epithelial differentiation of BM-MSCs by two types of defined induction media. The outcomes from the two media were different but, according to the results of CK3 and CK12 expressions, both media can induce the corneal epithelial differentiation.
In some experiments, combinations of differentiation methods were used and some stimulating conditions were applied in order to improve the results. In the differentiation of human adipose tissue-derived MSCs (AT-MSCs) by Bandeira et al. [8], induction media were used to initially induce AT-MSCs to become unspecific epithelial cells. Then, the initial induced cells were cultured as a cell sheet on fibrin gel before transplantation in rat LSCD model. The results revealed that initial induced AT-MSCs can undergo terminal differentiation in vivo into corneal epithelial cells (as shown by the expression of CK3 and CK12) and restored the corneal epithelium better than transplantation of undifferentiated AT-MSCs. In the differentiation of human BM-MSCs by Rohaina et al. [22], BM-MSCs were seeded on an amniotic membrane and co-cultured with mitomycin C-treated mouse fibroblast in an induction medium. Ten days after induction, the induced BM-MSCs formed cell sheet on the amniotic membrane and had gene expression of the determined markers (CK3, p63, C/EBPδ, β1-integrin) significantly higher than uninduced BM-MSCs. Moreover, Garzón et al. [23] can induce corneal epithelial differentiation of human Wharton’s jelly-derived MSCs (WJ-MSCs) to become corneal epithelium on their artificial cornea. Their artificial cornea was composed of two layers; stroma and epithelium. The stroma layer was fibrin–agarose gel seeded with human corneal keratocytes. The WJ-MSCs were seeded on the stroma layer. The constructs were submerged in an induction medium for 28 days. In such a condition, the differentiation of WJ-MSCs was induced by cytokines provided by the keratinocytes in the stroma layer and bioactive molecules in the induction medium. After that, the constructs were lifted up until the upper surface touched the air in order to induce stratification and terminal differentiation. By using this strategy, WJ-MSCs can differentiate into corneal epithelium and the obtained artificial cornea expressed marker profile similar to human cornea.

5. Corneal Epithelial Differentiation of Human PSCs

Both ESCs and iPSCs have pluripotent capacity to differentiate into many cell types. ESCs are isolated from inner cell mass of a blastocyst [17,122] while iPSCs are generated from embryonic or adult fibroblast cells [16,123]. These PSCs can be good candidates for treating LSCD. Transplantation of ESCs from animal and human successfully reconstructed ocular surface of host species [11,69,124,125,126]. Mouse ESCs could differentiate to be corneal epithelial-like cells (expressed CK12 and p63α), develop to form multilayer, heal porcine wounded cornea after transplantation, and maintain corneal marker expression [126]. Pax6 cDNA-transfected mouse ESCs showed expression of CK2, E-cadherin, and CD44 [69]. Moreover, they could expand to form three layers of epithelial cells on mouse damaged cornea [69]. Induced epithelial progenitor cells from mouse ESCs were successful generated and complete reconstructed mouse corneal surface after 24 h of transplantation [124]. Especially, cynomolgus monkey ESCs could be differentiated to be corneal epithelial-like cells which could form multiple cell layers on injured mouse cornea after transplantation [125]. Recently, transplantation of human ESCs has few publications, however, good results can be obtained. After six days of human ESCs transplantation onto human wounded cornea in vitro, these cells could expand on human Bowman’s membrane to produce one to four cell layers that expressed corneal markers (PAX6 and CK3) [127]. Noticeably, transplantation of epithelial cell sheet derived from human ESCs successfully recovered ocular surface of a LSCD rabbit and showed phenotype of limbal and corneal epithelial cells after eight weeks [11].
Although there is no publication about transplantation of iPSCs to treat LSCD eyes, these cells provide a valuable opportunity for vision therapy. Because iPSCs have similar differentiation capacity to ESCs but iPSCs can be generated from patients’ somatic cells so transplantation of autologous iPSCs can avoid immune rejection. Human iPSCs could be generate cornea organoids which expressed markers of adult corneal tissue [128] or displayed similar features of the developing cornea [129]. After explant cultures of minicorneas on denuded human amniotic membrane substrates for 10 days, the generated cell sheets expressed PAX6 and CK12 [128]. Human iPSC-organoid comprised of three cell types that express markers of corneal epithelium (CK3, p63), stroma (collagen type I and V), and endothelium (Collagen type VIII) [129].
To differentiate PSCs into corneal epithelial cells can use transgenic or non-transgenic methods. However, the transgenic method was only performed in mouse ESCs [69] while human PSCs only used non-transgenic method.
Non-transgenic method consists of the defined induction medium and CM. Several reports used CM from human limbal fibroblasts [25,130] or human limbal stromal cells [131] for induction of ESC/iPSC-derived corneal epithelial cells. Human limbal fibroblasts or stromal cells were isolated and expanded by subculture up to 10–15 passages [25,131]. These cells were treated with mitomycin C (MMC) for mitotic inactivation, then cultured in the corneal epithelial medium. The medium was changed daily and CM was collected every day until day even [25,130] or day 10 [131]. The detail component of corneal epithelial medium and the results are shown in Table 3. To collect a high population of derived corneal epithelial cells, the optimal time is ranged from one to two weeks depended on the cell types.
Differentiation of corneal epithelial cells from PSCs using the defined induction medium contains two or three steps (Table 3). Firstly, the ESCs/iPSCs are treated with induction media containing small molecules and/or growth factors involved in main signaling pathways that are necessary for corneal differentiation. The induction medium comprises defined and feeder-free medium (E6, RegES, mTeSR1, XF-Ko-SR) supplemented with small molecules (SB505124, IWP2 or IWR1, A83-01), bFGF with/without BMP4. Pluripotent cells will form embryonic bodies during suspension culture for about 4 days in the first step. After induction, pluripotent stem cells markers (Oct4, c-Myc, Nanog, Sox2) are significantly decreased while several transcription factors (PAX6, PITX2, BMP4, FOX1) regulated corneal development are increased [61]. Secondly, these cells are cultured in corneal epithelial medium (Cnt-30, CnT prime) or define-keratinocyte serum free medium (DKSFM) for further differentiation to corneal epithelial progenitor cells and maturation to corneal epithelial cells. Corneal epithelial progenitor cells show the markers of limbal stem cells such as p63, CK15, ABCG2, CK15, CK14 and the marker of proliferating cells (Ki67). Corneal epithelial cells express specific corneal markers (CK3, CK12) and other markers (CK15, CK14, and E-cadherin).
The efficiency of pluripotent stem cell-derived corneal epithelial cells is variable (Table 3). The highest efficiency is shown with 99% of cells positively expressed CK3 (H9 cell line) and 94% of cells expressed CK12 (RCM1 cell line) when the CM from limbal fibroblasts was used [130]. However, CK3 and CK12 protein expression results are very different from their mRNA expression results (CK3, CK12 mRNA expressed very low). Another report that also showed high efficiency of differentiation (70% CK12 positive, 30% CK3 positive) used RegES medium supplemented with two small molecules (IWP2, SB505124) and bFGF [61]. Moreover, they used a defined, serum-free, feeder free culture system that overcomes the disadvantage of using CM and co-culture system which had risks of pathogen transmission, immune response [132,133].
During the generation of pluripotent stem cell-derived corneal epithelial cells, collagen type IV is the most popular candidate for coating dish [25,61,83]. Collagen type IV, the main component of the basement membrane, is a form of extracellular matrix [134,135], supporting attachment and proliferation of limbal stem cells and corneal epithelial cells [25,136]. Another component of the basement membrane, laminin, is also used in combination with collagen type IV during differentiation into corneal epithelial cells [85,87].
Responsiveness of pluripotent stem cell lines are very different between cell lines [25,83,137]. Although the system for corneal epithelial cell generation is the same, the differentiated results are very variable. Some cell lines showed that they are not responsive for the induction of corneal epithelial cells (253G1, 201B7 [137], SB-Ad3 [83]). That maybe cause by the low activity of BMP signaling which could be improved by treatment with SB431542 [83].

6. Future Perspective

Even though the mechanism involving in the differentiation of stem cells into corneal epithelial cells has not yet been clearly clarified, many differentiation induction protocols have been proposed. Many publications reported the differentiation of stem cells into corneal epithelial cells but most of them are animal stem cell works. Only a limited number of publications studied the differentiation of human stem cells into corneal epithelial cells. There are differences in many aspects between the stem cells from animals and humans. Hence, the protocols developed from the studies on animal stem cells are probably not suitable to apply on human stem cells. Among the published reports of the corneal epithelial differentiation of human stem cells, various methods were used. Lack of comparison between the results obtained from each protocol causes unavailable of standard differentiation protocol. Therefore, the research to find good protocols suitable for each stem cell type is still required. This review concludes the published reports of the corneal epithelial differentiation of human MSCs and PSCs. According to the available reports, only small number of protocols have been investigated and the yields of successful differentiation are still not high. Therefore, protocols for inducing corneal epithelial differentiation have to be improved for better outcomes. Besides MSCs and PSCs, other kinds of human stem cells have also been investigated, especially the cells from ectoderm origin. Skin-derived precursor cells were induced corneal epithelial differentiation by a defined induction medium [138]. After the differentiation induction for 21 days, the result revealed incomplete differentiation because they expressed CK3 but did not express CK12. Oral mucosa epithelium is the source of adult cells which are widely used for corneal epithelium reconstruction [6,139,140,141,142]. The oral mucosa epithelial cells cultured on amniotic membranes offered satisfied outcomes after transplantation in patients in clinical trials. Umbilical cords from newborn babies are also the sources of cells for corneal epithelial regeneration. Umbilical cord lining epithelial cells cultured on human amniotic membrane expressed CK3 and CK12 [143] and can be used to treat patients with persistent epithelial defects [144].
Even though many kinds of human cells can be used for corneal epithelium regeneration, studies of corneal epithelial differentiation are still necessary to clarify the mechanism of differentiation. The obtained information from the investigations will provide useful knowledge in stem cell biology which is beneficial for medical applications in the future.

Author Contributions

Conceptualization, K.T. and H.T.N.; writing—original draft preparation, K.T. and H.T.N.; writing—review and editing, J.M., S.I., S.S., H.K. and M.K.-C.; supervision, R.P.; funding acquisition, R.P. All authors have read and agreed to the published version of the manuscript.

Funding

This review was supported by the Office of National Higher Education Science Research and Innovation Policy Council (NXPO) grant number B05F630042. Kasem Theerakittayakorn was supported by SUT Postgraduate research fellowships.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Palanker, D.; Blumenkranz, M.S. Chapter 39—Retinal Laser Therapy: Biophysical Basis and Applications. In Retina, 5th ed.; Ryan, S.J., Sadda, S.R., Hinton, D.R., Schachat, A.P., Sadda, S.R., Wilkinson, C.P., Wiedemann, P., Schachat, A.P., Eds.; W.B. Saunders: London, UK, 2013; pp. 746–760. [Google Scholar] [CrossRef]
  2. Chen, L.W.; Chen, Y.-M.; Lu, C.-J.; Chen, M.-Y.; Lin, S.-Y.; Hu, F.-R.; Chen, W.-L. Effect of air-lifting on the stemness, junctional protein formation, and cytokeratin expression of in vitro cultivated limbal epithelial cell sheets. Taiwan J. Ophthalmol. 2017, 7, 205–212. [Google Scholar] [CrossRef] [PubMed]
  3. Nakamura, T.; Endo, K.-I.; Cooper, L.J.; Fullwood, N.J.; Tanifuji, N.; Tsuzuki, M.; Koizumi, N.; Inatomi, T.; Sano, Y.; Kinoshita, S. The Successful Culture and Autologous Transplantation of Rabbit Oral Mucosal Epithelial Cells on Amniotic Membrane. Investig. Ophthalmol. Vis. Sci. 2003, 44, 106–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Tsai, R.J.; Li, L.M.; Chen, J.K. Reconstruction of damaged corneas by transplantation of autologous limbal epithelial cells. N. Eng. J. Med. 2000, 343, 86–93. [Google Scholar] [CrossRef] [PubMed]
  5. Mathews, S.; Prasad, T.; Venugopal, B.; Palakkan, A.A.; Anil Kumar, P.R.; Kumary, T.V. Standardizing transdifferentiation of rabbit bone marrow mesenchymal stem cells to corneal lineage by simulating corneo-limbal cues. J. Stem Cell Res. Med. 2017, 2, 1–10. [Google Scholar] [CrossRef]
  6. Samoila, O.; Gocan, D. Clinical Outcomes from Cultivated Allogenic Stem Cells vs. Oral Mucosa Epithelial Transplants in Total Bilateral Stem Cells Deficiency. Front. Med. 2020, 7. [Google Scholar] [CrossRef] [Green Version]
  7. 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]
  8. Bandeira, F.; Goh, T.-W.; Setiawan, M.; Yam, G.H.-F.; Mehta, J.S. Cellular therapy of corneal epithelial defect by adipose mesenchymal stem cell-derived epithelial progenitors. Stem Cell Res. Ther. 2020, 11, 14. [Google Scholar] [CrossRef] [Green Version]
  9. Da Mata Martins, T.M.; da Silva Cunha, P.; Rodrigues, M.A.; de Carvalho, J.L.; de Souza, J.E.; de Carvalho Oliveira, J.A.; Gomes, D.A.; de Goes, A.M. Epithelial basement membrane of human decellularized cornea as a suitable substrate for differentiation of embryonic stem cells into corneal epithelial-like cells. Mater. Sci. Eng. C 2020, 116. [Google Scholar] [CrossRef]
  10. Zhang, P.; Ma, X.-y.; Huang, D.-t.; Yang, X.-y. The capacity of goat epidermal adult stem cells to reconstruct the damaged ocular surface of total LSCD and activate corneal genetic programs. J. Mol. Histol. 2020, 51, 277–286. [Google Scholar] [CrossRef]
  11. He, J.; Ou, S.; Ren, J.; Sun, H.; He, X.; Zhao, Z.; Wu, H.; Qu, Y.; Liu, T.; Jeyalatha, V.; et al. Tissue engineered corneal epithelium derived from clinical-grade human embryonic stem cells. Ocul. Surf. 2020, 18, 672–680. [Google Scholar] [CrossRef]
  12. Le-Bel, G.; Guérin, L.-P.; Carrier, P.; Mouriaux, F.; Germain, L.; Guérin, S.L.; Bazin, R. Grafting of an autologous tissue-engineered human corneal epithelium to a patient with limbal stem cell deficiency (LSCD). Am. J. Ophthalmol. Case Rep. 2019, 15, 100532. [Google Scholar] [CrossRef] [PubMed]
  13. Marquez-Curtis, L.A.; Janowska-Wieczorek, A.; McGann, L.E.; Elliott, J.A.W. Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects. Cryobiology 2015, 71, 181–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Elahi, K.C.; Klein, G.; Avci-Adali, M.; Sievert, K.D.; MacNeil, S.; Aicher, W.K. Human Mesenchymal Stromal Cells from Different Sources Diverge in Their Expression of Cell Surface Proteins and Display Distinct Differentiation Patterns. Stem Cells Int. 2016, 2016, 5646384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Payushina, O.V.; Domaratskaya, E.I.; Starostin, V.I. Mesenchymal stem cells: Sources, phenotype, and differentiation potential. Biol. Bull. 2006, 33, 2–18. [Google Scholar] [CrossRef]
  16. Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Thomson, J.A.; Itskovitz-Eldor, J.; Shapiro, S.S.; Waknitz, M.A.; Swiergiel, J.J.; Marshall, V.S.; Jones, J.M. Embryonic stem cell lines derived from human blastocysts. Science 1998, 282, 1145–1147. [Google Scholar] [CrossRef] [Green Version]
  18. Prescott, C. The business of exploiting induced pluripotent stem cells. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2011, 366, 2323–2328. [Google Scholar] [CrossRef] [Green Version]
  19. D’Antonio, M.; Woodruff, G.; Nathanson, J.L.; D’Antonio-Chronowska, A.; Arias, A.; Matsui, H.; Williams, R.; Herrera, C.; Reyna, S.M.; Yeo, G.W.; et al. High-Throughput and Cost-Effective Characterization of Induced Pluripotent Stem Cells. Stem Cell Rep. 2017, 8, 1101–1111. [Google Scholar] [CrossRef]
  20. Katikireddy, K.R.; Dana, R.; Jurkunas, U.V. Differentiation Potential of Limbal Fibroblasts and Bone Marrow Mesenchymal Stem Cells to Corneal Epithelial Cells. Stem Cells 2014, 32, 717–729. [Google Scholar] [CrossRef]
  21. Nieto-Nicolau, N.; Martín-Antonio, B.; Müller-Sánchez, C.; Casaroli-Marano, R.P. In vitro potential of human mesenchymal stem cells for corneal epithelial regeneration. Regen. Med. 2020, 15, 1409–1426. [Google Scholar] [CrossRef]
  22. Rohaina, C.M.; Then, K.Y.; Ng, A.M.H.; Wan Abdul Halim, W.H.; Zahidin, A.Z.M.; Saim, A.; Idrus, R.B.H. Reconstruction of limbal stem cell deficient corneal surface with induced human bone marrow mesenchymal stem cells on amniotic membrane. Transl. Res. 2014, 163, 200–210. [Google Scholar] [CrossRef] [PubMed]
  23. Garzón, I.; Martín-Piedra, M.A.; Alfonso-Rodríguez, C.; González-Andrades, M.; Carriel, V.; Martínez-Gómez, C.; Campos, A.; Alaminos, M. Generation of a Biomimetic Human Artificial Cornea Model Using Wharton’s Jelly Mesenchymal Stem Cells. Investig. Ophthalmol. Vis. Sci. 2014, 55, 4073–4083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Soleimanifar, F.; Mortazavi, Y.; Nadri, S.; Islami, M.; Vakilian, S. Coculture of conjunctiva derived mesenchymal stem cells (CJMSCs) and corneal epithelial cells to reconstruct the corneal epithelium. Biologicals 2018, 54, 39–43. [Google Scholar] [CrossRef] [PubMed]
  25. Ahmad, S.; Stewart, 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]
  26. Ginis, I.; Luo, Y.; Miura, T.; Thies, S.; Brandenberger, R.; Gerecht-Nir, S.; Amit, M.; Hoke, A.; Carpenter, M.K.; Itskovitz-Eldor, J.; et al. Differences between human and mouse embryonic stem cells. Dev. Biol. 2004, 269, 360–380. [Google Scholar] [CrossRef] [Green Version]
  27. Hillmann, A.; Ahrberg, A.B.; Brehm, W.; Heller, S.; Josten, C.; Paebst, F.; Burk, J. Comparative Characterization of Human and Equine Mesenchymal Stromal Cells: A Basis for Translational Studies in the Equine Model. Cell Transpl. 2016, 25, 109–124. [Google Scholar] [CrossRef] [Green Version]
  28. Schnerch, A.; Cerdan, C.; Bhatia, M. Distinguishing between mouse and human pluripotent stem cell regulation: The best laid plans of mice and men. Stem Cells 2010, 28, 419–430. [Google Scholar] [CrossRef]
  29. Bottagisio, M.; Lovati, A.B.; Lopa, S.; Moretti, M. Osteogenic Differentiation of Human and Ovine Bone Marrow Stromal Cells in response to β-Glycerophosphate and Monosodium Phosphate. Cell. Reprogram. 2015, 17, 235–242. [Google Scholar] [CrossRef]
  30. Kalaszczynska, I.; Ruminski, S.; Platek, A.E.; Bissenik, I.; Zakrzewski, P.; Noszczyk, M.; Lewandowska-Szumiel, M. Substantial differences between human and ovine mesenchymal stem cells in response to osteogenic media: How to explain and how to manage? Biores. Open Access 2013, 2, 356–363. [Google Scholar] [CrossRef]
  31. Arnalich-Montiel, F.; Pastor, S.; Blazquez-Martinez, A.; Fernandez-Delgado, J.; Nistal, M.; Alio, J.L.; De Miguel, M.P. Adipose-Derived Stem Cells Are a Source for Cell Therapy of the Corneal Stroma. Stem Cells 2008, 26, 570–579. [Google Scholar] [CrossRef]
  32. Oliva, J.; Bardag-Gorce, F.; Niihara, Y. Clinical Trials of Limbal Stem Cell Deficiency Treated with Oral Mucosal Epithelial Cells. Int. J. Mol. Sci. 2020, 21, 411. [Google Scholar] [CrossRef] [Green Version]
  33. Movahedan, A.; Majdi, M.; Afsharkhamseh, N.; Sagha, H.M.; Saadat, N.S.; Shalileh, K.; Milani, B.Y.; Ying, H.; Djalilian, A.R. Notch Inhibition during Corneal Epithelial Wound Healing Promotes Migration. Investig. Ophthalmol. Vis. Sci. 2012, 53, 7476–7483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sridhar, M.S. Anatomy of cornea and ocular surface. Indian J. Ophthalmol. 2018, 66, 190–194. [Google Scholar] [CrossRef] [PubMed]
  35. Joseph, A.; Powell-Richards, A.O.R.; Shanmuganathan, V.A.; Dua, H.S. Epithelial cell characteristics of cultured human limbal explants. Br. J. Ophthalmol. 2004, 88, 393–398. [Google Scholar] [CrossRef] [Green Version]
  36. Mariappan, I.; Maddileti, S.; Savy, S.; Tiwari, S.; Gaddipati, S.; Fatima, A.; Sangwan, V.S.; Balasubramanian, D.; Vemuganti, G.K. In vitro culture and expansion of human limbal epithelial cells. Nat. Protoc. 2010, 5, 1470. [Google Scholar] [CrossRef] [PubMed]
  37. Reinstein Merjava, S.; Neuwirth, A.; Tanzerova, M.; Jirsova, K. The spectrum of cytokeratins expressed in the adult human cornea, limbus and perilimbal conjunctiva. Histol. Histopathol. 2011, 26, 323–331. [Google Scholar] [CrossRef]
  38. Wijnholds, J. “Basal Cell Migration” in Regeneration of the Corneal Wound-Bed. Stem Cell Rep. 2019, 12, 3–5. [Google Scholar] [CrossRef] [PubMed]
  39. Mort, R.L.; Douvaras, P.; Morley, S.D.; Dorà, N.; Hill, R.E.; Collinson, J.M.; West, J.D. Stem cells and corneal epithelial maintenance: Insights from the mouse and other animal models. Results Probl. Cell Differ. 2012, 55, 357–394. [Google Scholar] [CrossRef] [Green Version]
  40. Beebe, D.C.; Masters, B.R. Cell lineage and the differentiation of corneal epithelial cells. Investig. Ophthalmol. Vis. Sci. 1996, 37, 1815–1825. [Google Scholar]
  41. Swamynathan, S.K. Ocular surface development and gene expression. J. Ophthalmol. 2013, 2013, 103947. [Google Scholar] [CrossRef] [Green Version]
  42. Yazdanpanah, G.; Jabbehdari, S.; Djalilian, A.R. Limbal and corneal epithelial homeostasis. Curr. Opin. Ophthalmol. 2017, 28, 348–354. [Google Scholar] [CrossRef]
  43. López-Paniagua, M.; Nieto-Miguel, T.; de la Mata, A.; Dziasko, M.; Galindo, S.; Rey, E.; Herreras, J.M.; Corrales, R.M.; Daniels, J.T.; Calonge, M. Comparison of functional limbal epithelial stem cell isolation methods. Exp. Eye Res. 2016, 146, 83–94. [Google Scholar] [CrossRef] [PubMed]
  44. Kruse, F.E.; Tseng, S.C. Growth factors modulate clonal growth and differentiation of cultured rabbit limbal and corneal epithelium. Investig. Ophthalmol. Vis. Sci. 1993, 34, 1963–1976. [Google Scholar]
  45. Kruse, F.E. Stem cells and corneal epithelial regeneration. Eye 1994, 8, 170–183. [Google Scholar] [CrossRef]
  46. Moll, R.; Franke, W.W.; Schiller, D.L.; Geiger, B.; Krepler, R. The catalog of human cytokeratins: Patterns of expression in normal epithelia, tumors and cultured cells. Cell 1982, 31, 11–24. [Google Scholar] [CrossRef]
  47. Schermer, A.; Galvin, S.; Sun, T.T. Differentiation-related expression of a major 64K corneal keratin in vivo and in culture suggests limbal location of corneal epithelial stem cells. J. Cell Biol. 1986, 103, 49–62. [Google Scholar] [CrossRef] [PubMed]
  48. Moll, R.; Divo, M.; Langbein, L. The human keratins: Biology and pathology. Histochem. Cell Biol. 2008, 129, 705–733. [Google Scholar] [CrossRef] [Green Version]
  49. Martínez García de la Torre, R.A.; Nieto-Nicolau, N.; Morales-Pastor, A.; Casaroli-Marano, R.P. Determination of the Culture Time Point to Induce Corneal Epithelial Differentiation in Induced Pluripotent Stem Cells. Transplant. Proc. 2017, 49, 2292–2295. [Google Scholar] [CrossRef]
  50. Harkin, D.G.; Foyn, L.; Bray, L.J.; Sutherland, A.J.; Li, F.J.; Cronin, B.G. Concise Reviews: Can Mesenchymal Stromal Cells Differentiate into Corneal Cells? A Systematic Review of Published Data. Stem Cells 2015, 33, 785–791. [Google Scholar] [CrossRef] [Green Version]
  51. Adhikary, G.; Crish, J.; Lass, J.; Eckert, R.L. Regulation of Involucrin Expression in Normal Human Corneal Epithelial Cells: A Role for Activator Protein One. Investig. Ophthalmol. Vis. Sci. 2004, 45, 1080–1087. [Google Scholar] [CrossRef] [Green Version]
  52. Adhikary, G.; Crish, J.F.; Gopalakrishnan, R.; Bone, F.; Eckert, R.L. Involucrin Expression in the Corneal Epithelium: An Essential Role for Sp1 Transcription Factors. Investig. Ophthalmol. Vis. Sci. 2005, 46, 3109–3120. [Google Scholar] [CrossRef] [PubMed]
  53. Khoh-Reiter, S.; Jessen, B.A. Evaluation of the cytotoxic effects of ophthalmic solutions containing benzalkonium chloride on corneal epithelium using an organotypic 3-D model. BMC Ophthalmol. 2009, 9, 5. [Google Scholar] [CrossRef] [Green Version]
  54. Guo, Z.H.; Zhang, W.; Jia, Y.Y.S.; Liu, Q.X.; Li, Z.F.; Lin, J.S. An Insight into the Difficulties in the Discovery of Specific Biomarkers of Limbal Stem Cells. Int. J. Mol. Sci. 2018, 19, 1982. [Google Scholar] [CrossRef] [Green Version]
  55. Yoon, J.J.; Ismail, S.; Sherwin, T. Limbal stem cells: Central concepts of corneal epithelial homeostasis. World J. Stem Cells 2014, 6, 391–403. [Google Scholar] [CrossRef] [PubMed]
  56. Arkell, R.M.; Tam, P.P.L. Initiating head development in mouse embryos: Integrating signalling and transcriptional activity. Open Biol. 2012, 2, 120030. [Google Scholar] [CrossRef] [Green Version]
  57. Gage, P.J.; Zacharias, A.L. Signaling “cross-talk” is integrated by transcription factors in the development of the anterior segment in the eye. Dev. Dyn. 2009, 238, 2149–2162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Fuhrmann, S. Wnt signaling in eye organogenesis. Organogenesis 2008, 4, 60–67. [Google Scholar] [CrossRef] [Green Version]
  59. Lian, X.; Selekman, J.; Bao, X.; Hsiao, C.; Zhu, K.; Palecek, S.P. A Small Molecule Inhibitor of Src Family Kinases Promotes Simple Epithelial Differentiation of Human Pluripotent Stem Cells. PLoS ONE 2013, 8, e60016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Zhang, Y.; Yeh, L.-K.; Zhang, S.; Call, M.; Yuan, Y.; Yasunaga, M.; Kao, W.W.Y.; Liu, C.-Y. Wnt/β-catenin signaling modulates corneal epithelium stratification via inhibition of Bmp4 during mouse development. Development 2015, 142, 3383–3393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. 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, 2, 219–231. [Google Scholar] [CrossRef] [Green Version]
  62. Glinka, A.; Wu, W.; Delius, H.; Monaghan, A.P.; Blumenstock, C.; Niehrs, C. Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction. Nature 1998, 391, 357–362. [Google Scholar] [CrossRef] [PubMed]
  63. Machon, O.; Kreslova, J.; Ruzickova, J.; Vacik, T.; Klimova, L.; Fujimura, N.; Lachova, J.; Kozmik, Z. Lens morphogenesis is dependent on Pax6-mediated inhibition of the canonical Wnt/beta-catenin signaling in the lens surface ectoderm. Genesis 2010, 48, 86–95. [Google Scholar] [CrossRef] [PubMed]
  64. Gage, P.J.; Qian, M.; Wu, D.; Rosenberg, K.I. The canonical Wnt signaling antagonist DKK2 is an essential effector of PITX2 function during normal eye development. Dev. Biol. 2008, 317, 310–324. [Google Scholar] [CrossRef] [Green Version]
  65. Forsdahl, S.; Kiselev, Y.; Hogseth, R.; Mjelle, J.E.; Mikkola, I. Pax6 Regulates the Expression of Dkk3 in Murine and Human Cell Lines, and Altered Responses to Wnt Signaling Are Shown in FlpIn-3T3 Cells Stably Expressing Either the Pax6 or the Pax6(5a) Isoform. PLoS ONE 2014, 9, e102559. [Google Scholar] [CrossRef] [Green Version]
  66. Tsukiyama, T.; Fukui, A.; Terai, S.; Fujioka, Y.; Shinada, K.; Takahashi, H.; Yamaguchi, T.P.; Ohba, Y.; Hatakeyama, S. Molecular Role of RNF43 in Canonical and Noncanonical Wnt Signaling. Mol. Cell. Biol. 2015, 35, 2007–2023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Madan, B.; Virshup, D.M. Targeting Wnts at the source--new mechanisms, new biomarkers, new drugs. Mol. Cancer Ther. 2015, 14, 1087–1094. [Google Scholar] [CrossRef] [Green Version]
  68. Gan, Q.; Lee, A.; Suzuki, R.; Yamagami, T.; Stokes, A.; Nguyen, B.C.; Pleasure, D.; Wang, J.; Chen, H.-W.; Zhou, C.J. Pax6 mediates ß-catenin signaling for self-renewal and neurogenesis by neocortical radial glial stem cells. Stem Cells 2014, 32, 45–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. 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]
  70. Sun, J.; Liu, W.-H.; Deng, F.-M.; Luo, Y.-H.; Wen, K.; Zhang, H.; Liu, H.-R.; Wu, J.; Su, B.-Y.; Liu, Y.-L. Differentiation of rat adipose-derived mesenchymal stem cells into corneal-like epithelial cells driven by PAX6. Exp. Ther. Med. 2018, 15, 1424–1432. [Google Scholar] [CrossRef]
  71. Sasamoto, Y.; Hayashi, R.; Park, S.-J.; Saito-Adachi, M.; Suzuki, Y.; Kawasaki, S.; Quantock, A.J.; Nakai, K.; Tsujikawa, M.; Nishida, K. PAX6 Isoforms, along with Reprogramming Factors, Differentially Regulate the Induction of Cornea-specific Genes. Sci. Rep. 2016, 6, 20807. [Google Scholar] [CrossRef] [Green Version]
  72. Heavner, W.; Pevny, L. Eye development and retinogenesis. Cold Spring Harb. Perspect. Biol. 2012, 4, a008391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Chen, B.; Dodge, M.E.; Tang, W.; Lu, J.; Ma, Z.; Fan, C.-W.; Wei, S.; Hao, W.; Kilgore, J.; Williams, N.S.; et al. Small molecule-mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer. Nat. Chem. Biol. 2009, 5, 100–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Lee, H.J.; Wolosin, J.M.; Chung, S.-H. Divergent effects of Wnt/β-catenin signaling modifiers on the preservation of human limbal epithelial progenitors according to culture condition. Sci. Rep. 2017, 7, 15241. [Google Scholar] [CrossRef] [PubMed]
  75. Huang, H.; He, X. Wnt/beta-catenin signaling: New (and old) players and new insights. Curr. Opin. Cell Biol. 2008, 20, 119–125. [Google Scholar] [CrossRef] [Green Version]
  76. Yang, J.; Park, J.W.; Zheng, D.; Xu, R.-H. Universal Corneal Epithelial-Like Cells Derived from Human Embryonic Stem Cells for Cellularization of a Corneal Scaffold. Transl. Vis. Sci. Technol. 2018, 7, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Hamilton, P.W.; Sun, Y.; Henry, J.J. Lens regeneration from the cornea requires suppression of Wnt/β-catenin signaling. Exp. Eye Res. 2016, 145, 206–215. [Google Scholar] [CrossRef] [Green Version]
  78. Kahata, K.; Dadras, M.S.; Moustakas, A. TGF-beta Family Signaling in Epithelial Differentiation and Epithelial-Mesenchymal Transition. Cold Spring Harb. Perspect. Biol. 2018, 10. [Google Scholar] [CrossRef] [Green Version]
  79. Inman, G.J.; Nicolás, F.J.; Callahan, J.F.; Harling, J.D.; Gaster, L.M.; Reith, A.D.; Laping, N.J.; Hill, C.S. SB-431542 Is a Potent and Specific Inhibitor of Transforming Growth Factor-β Superfamily Type I Activin Receptor-Like Kinase (ALK) Receptors ALK4, ALK5, and ALK7. Mol. Pharmacol. 2002, 62, 65–74. [Google Scholar] [CrossRef]
  80. DaCosta Byfield, S.; Major, C.; Laping, N.J.; Roberts, A.B. SB-505124 is a selective inhibitor of transforming growth factor-beta type I receptors ALK4, ALK5, and ALK7. Mol. Pharm. 2004, 65, 744–752. [Google Scholar] [CrossRef]
  81. Tojo, M.; Hamashima, Y.; Hanyu, A.; Kajimoto, T.; Saitoh, M.; Miyazono, K.; Node, M.; Imamura, T. The ALK-5 inhibitor A-83-01 inhibits Smad signaling and epithelial-to-mesenchymal transition by transforming growth factor-β. Cancer Sci. 2005, 96, 791–800. [Google Scholar] [CrossRef]
  82. Wang, D.; Wang, E.; Liu, K.; Xia, C.-h.; Li, S.; Gong, X. Roles of TGFβ and FGF signals during growth and differentiation of mouse lens epithelial cell in vitro. Sci. Rep. 2017, 7, 7274. [Google Scholar] [CrossRef] [PubMed]
  83. Kamarudin, T.A.; Bojic, S.; Collin, J.; Yu, M.; Alharthi, S.; Buck, H.; Shortt, A.; Armstrong, L.; Figueiredo, F.C.; Lako, M. Differences in the Activity of Endogenous Bone Morphogenetic Protein Signaling Impact on the Ability of Induced Pluripotent Stem Cells to Differentiate to Corneal Epithelial-Like Cells. Stem Cells 2018, 36, 337–348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Chang, H.M.; Cheng, J.C.; Fang, L.; Qiu, X.; Klausen, C.; Taylor, E.L.; Leung, P.C. Recombinant BMP4 and BMP7 downregulate pentraxin 3 in human granulosa cells. J. Clin. Endocrinol. Metab. 2015, 100, E365–E374. [Google Scholar] [CrossRef] [Green Version]
  85. Hongisto, H.; Ilmarinen, T.; Vattulainen, M.; Mikhailova, A.; Skottman, H. Xeno- and feeder-free differentiation of human pluripotent stem cells to two distinct ocular epithelial cell types using simple modifications of one method. Stem Cell Res. Ther. 2017, 8, 291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Hongisto, H.; Vattulainen, M.; Ilmarinen, T.; Mikhailova, A.; Skottman, H. Efficient and Scalable Directed Differentiation of Clinically Compatible Corneal Limbal Epithelial Stem Cells from Human Pluripotent Stem Cells. J. Vis. Exp. 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Vattulainen, M.; Ilmarinen, T.; Koivusalo, L.; Viiri, K.; Hongisto, H.; Skottman, H. Modulation of Wnt/BMP pathways during corneal differentiation of hPSC maintains ABCG2-positive LSC population that demonstrates increased regenerative potential. Stem Cell Res. Ther. 2019, 10, 236. [Google Scholar] [CrossRef]
  88. Fitzsimmons, R.E.B.; Mazurek, M.S.; Soos, A.; Simmons, C.A. Mesenchymal Stromal/Stem Cells in Regenerative Medicine and Tissue Engineering. Stem Cells Int. 2018, 2018, 8031718. [Google Scholar] [CrossRef]
  89. Najar, M.; Fahmi, H. Of Mesenchymal Stem/Stromal Cells and Osteoarthritis: Time to Merge the Latest Breakthroughs. Stem Cell Rev. Rep. 2020, 16, 1016–1018. [Google Scholar] [CrossRef]
  90. Kim, H.J.; Park, J.-S. Usage of Human Mesenchymal Stem Cells in Cell-based Therapy: Advantages and Disadvantages. Dev. Reprod. 2017, 21, 1–10. [Google Scholar] [CrossRef] [Green Version]
  91. Musiał-Wysocka, A.; Kot, M.; Majka, M. The Pros and Cons of Mesenchymal Stem Cell-Based Therapies. Cell Transplant. 2019, 28, 801–812. [Google Scholar] [CrossRef] [Green Version]
  92. Squillaro, T.; Peluso, G.; Galderisi, U. Clinical Trials With Mesenchymal Stem Cells: An Update. Cell Transplant. 2016, 25, 829–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Andrzejewska, A.; Lukomska, B.; Janowski, M. Concise Review: Mesenchymal Stem Cells: From Roots to Boost. Stem Cells 2019, 37, 855–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Viswanathan, S.; Shi, Y.; Galipeau, J.; Krampera, M.; Leblanc, K.; Martin, I.; Nolta, J.; Phinney, D.G.; Sensebe, L. Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT®) Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy 2019, 21, 1019–1024. [Google Scholar] [CrossRef] [PubMed]
  95. Miwa, H.; Era, T. Tracing the destiny of mesenchymal stem cells from embryo to adult bone marrow and white adipose tissue via Pdgfrα expression. Development 2018, 145, dev155879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Jeon, B.-G.; Jang, S.-J.; Park, J.-S.; Subbarao, R.B.; Jeong, G.-J.; Park, B.-W.; Rho, G.-J. Differentiation potential of mesenchymal stem cells isolated from human dental tissues into non-mesodermal lineage. Anim. Cells Syst. 2015, 19, 321–331. [Google Scholar] [CrossRef] [Green Version]
  97. Azandeh, S.; Mohammad Gharravi, A.; Orazizadeh, M.; Khodadi, A.; Hashemi Tabar, M. Improvement of mesenchymal stem cell differentiation into the endoderm lineage by four step sequential method in biocompatible biomaterial. Bioimpacts 2016, 6, 9–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Jadalannagari, S.; Aljitawi, O.S. Ectodermal Differentiation of Wharton’s Jelly Mesenchymal Stem Cells for Tissue Engineering and Regenerative Medicine Applications. Tissue Eng. Part B Rev. 2014, 21, 314–322. [Google Scholar] [CrossRef]
  99. Jadalannagari, S.; Berry, A.M.; Hopkins, R.A.; Bhavsar, D.; Aljitawi, O.S. Potential mechanisms underlying ectodermal differentiation of Wharton’s jelly mesenchymal stem cells. Biochem. Biophys. Res. Commun. 2016, 478, 831–837. [Google Scholar] [CrossRef]
  100. Sacchetti, M.; Rama, P.; Bruscolini, A.; Lambiase, A. Limbal Stem Cell Transplantation: Clinical Results, Limits, and Perspectives. Stem Cells Int. 2018, 2018, 8086269. [Google Scholar] [CrossRef] [Green Version]
  101. Guo, T.; Wang, W.; Zhang, J.; Chen, X.; Li, B.; Li, L. Experimental study on repairing damage of corneal surface by mesenchymal stem cells transplantation. Chin. J. Ophthalmol. 2006, 42, 246–250. [Google Scholar]
  102. Monteiro, B.G.; Serafim, R.C.; Melo, G.B.; Silva, M.C.P.; Lizier, N.F.; Maranduba, C.M.C.; Smith, R.L.; Kerkis, A.; Cerruti, H.; Gomes, J.A.P.; 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]
  103. Gomes, J.Á.P.; 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. Investig. Ophthalmol. Vis. Sci. 2010, 51, 1408–1414. [Google Scholar] [CrossRef]
  104. Lin, K.-J.; Loi, M.-X.; Lien, G.-S.; Cheng, C.-F.; Pao, H.-Y.; Chang, Y.-C.; Ji, A.T.-Q.; Ho, J.H.-C. Topical administration of orbital fat-derived stem cells promotes corneal tissue regeneration. Stem Cell Res. Ther. 2013, 4, 72. [Google Scholar] [CrossRef] [Green Version]
  105. Zeppieri, M.; Salvetat, M.; Beltrami, A.; Cesselli, D.; Bergamin, N.; Russo, R.; Cavaliere, F.; Varano, G.; Alcalde, I.; Merayo-Lloves, J.; et al. Human Adipose-Derived Stem Cells for the Treatment of Chemically Burned Rat Cornea: Preliminary Results. Curr. Eye Res. 2013, 38. [Google Scholar] [CrossRef] [PubMed]
  106. Galindo, S.; Herreras, J.M.; López-Paniagua, M.; Rey, E.; de la Mata, A.; Plata-Cordero, M.; Calonge, M.; Nieto-Miguel, T. Therapeutic Effect of Human Adipose Tissue-Derived Mesenchymal Stem Cells in Experimental Corneal Failure Due to Limbal Stem Cell Niche Damage. Stem Cells 2017, 35, 2160–2174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Reinach, P.; Pokorny, K. The corneal epithelium: Clinical relevance of cytokine-mediated responses to maintenance of corneal health. Arq. Bras. Oftalmol. 2008, 71, 80–86. [Google Scholar] [CrossRef] [Green Version]
  108. Kruse, F.E.; Tseng, S.C. Retinoic acid regulates clonal growth and differentiation of cultured limbal and peripheral corneal epithelium. Investig. Ophthalmol. Vis. Sci. 1994, 35, 2405–2420. [Google Scholar]
  109. Yu, F.-S.X.; Yin, J.; Xu, K.; Huang, J. Growth factors and corneal epithelial wound healing. Brain Res. Bull. 2010, 81, 229–235. [Google Scholar] [CrossRef] [Green Version]
  110. Xue, G.; Han, X.; Ma, X.; Wu, H.; Qin, Y.; Liu, J.; Hu, Y.; Hong, Y.; Hou, Y. Effect of Microenvironment on Differentiation of Human Umbilical Cord Mesenchymal Stem Cells into Hepatocytes In Vitro and In Vivo. BioMed Res. Int. 2016, 2016, 8916534. [Google Scholar] [CrossRef] [Green Version]
  111. Keung, A.J.; Kumar, S.; Schaffer, D.V. Presentation counts: Microenvironmental regulation of stem cells by biophysical and material cues. Annu. Rev. Cell Dev. Biol. 2010, 26, 533–556. [Google Scholar] [CrossRef]
  112. Wan, P.-X.; Wang, B.-W.; Wang, Z.-C. Importance of the stem cell microenvironment for ophthalmological cell-based therapy. World J. Stem Cells 2015, 7, 448–460. [Google Scholar] [CrossRef] [PubMed]
  113. Steens, J.; Klein, D. Current Strategies to Generate Human Mesenchymal Stem Cells In Vitro. Stem Cells Int. 2018, 2018, 6726185. [Google Scholar] [CrossRef] [PubMed]
  114. Abdal Dayem, A.; Lee, S.B.; Kim, K.; Lim, K.M.; Jeon, T.-I.; Seok, J.; Cho, S.-G. Production of Mesenchymal Stem Cells Through Stem Cell Reprogramming. Int. J. Mol. Sci. 2019, 20, 1922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. 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]
  116. Raj, D.K.; Kumary, T.V.; Anil Kumar, P.R. Development of corneal epithelial cell sheet construct from trans-differentiated bone marrow mesenchymal stem cells. Adv. Tissue Eng. Regen. Med. 2018, 4, 127–135. [Google Scholar] [CrossRef]
  117. Venugopal, B.; Shenoy, S.J.; Mohan, S.; Anil Kumar, P.R.; Kumary, T.V. Bioengineered corneal epithelial cell sheet from mesenchymal stem cells-A functional alternative to limbal stem cells for ocular surface reconstruction. J. Biomed. Mater. Res. Part B Appl. Biomater. 2020, 108, 1033–1045. [Google Scholar] [CrossRef]
  118. 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]
  119. Sikora, B.; Skubis-Sikora, A.; Kimsa-Furdzik, M.; Ciszek, W.; Kostrzewski, M.; Stojko, J.; Mazurek, U.; Gola, J. Adipose-derived stem cells undergo differentiation after co-culture with porcine limbal epithelial stem cells. Stem Cell Res. 2019, 41, 101609. [Google Scholar] [CrossRef]
  120. Tsai, C.L.; Chuang, P.C.; Kuo, H.K.; Chen, Y.H.; Su, W.H.; Wu, P.C. Differentiation of Stem Cells From Human Exfoliated Deciduous Teeth Toward a Phenotype of Corneal Epithelium In Vitro. Cornea 2015, 34, 1471–1477. [Google Scholar] [CrossRef]
  121. Soleimanifar, F.; Mortazavi, Y.; Nadri, S.; Soleimani, M. Conjunctiva derived mesenchymal stem cell (CJMSCs) as a potential platform for differentiation into corneal epithelial cells on bioengineered electrospun scaffolds. J. Biomed. Mater. Res. Part A 2017, 105, 2703–2711. [Google Scholar] [CrossRef]
  122. Ding, Y.; Gan, Y.; Feng, J.B.; Qi, S.H.; Li, R.M.; Li, D.S. Efficient isolation inner cell mass from blastocysts by improved microsurgical technique. Cell Res. 2008, 18, S39. [Google Scholar] [CrossRef] [Green Version]
  123. 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]
  124. 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. Investig. Ophthalmol. Vis. Sci. 2004, 45, 4320–4326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Kumagai, Y.; Kurokawa, M.S.; Ueno, H.; Kayama, M.; Tsubota, K.; Nakatsuji, N.; Nito, S.; Ueno, S.; Suzuki, N. Induction of Corneal Epithelium-Like Cells From Cynomorgus Monkey Embrionic Stem Cells and Their Experimental Transplantation to Damaged Cornea. Investig. Ophthalmol. Vis. Sci. 2007, 48, 432–438. [Google Scholar]
  126. 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]
  127. Hanson, C.; Hardarson, T.; Ellerström, 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] [Green Version]
  128. Susaimanickam, P.J.; Maddileti, S.; Pulimamidi, V.K.; Boyinpally, S.R.; Naik, R.R.; Naik, M.N.; Reddy, G.B.; Sangwan, V.S.; Mariappan, I. Generating minicorneal organoids from human induced pluripotent stem cells. Development 2017, 144, 2338. [Google Scholar] [CrossRef] [Green Version]
  129. Foster, J.W.; Wahlin, K.; Adams, S.M.; Birk, D.E.; Zack, D.J.; Chakravarti, S. Cornea organoids from human induced pluripotent stem cells. Sci. Rep. 2017, 7, 41286. [Google Scholar] [CrossRef]
  130. Brzeszczynska, J.; Samuel, K.; Greenhough, S.; Ramaesh, K.; Dhillon, B.; Hay, D.C.; Ross, J.A. Differentiation and molecular profiling of human embryonic stem cell-derived corneal epithelial cells. Int. J. Mol. Med. 2014, 33, 1597–1606. [Google Scholar] [CrossRef] [Green Version]
  131. Cieslar-Pobuda, A.; Rafat, M.; Knoflach, V.; Skonieczna, M.; Hudecki, A.; Malecki, A.; Urasinska, E.; Ghavami, S.; Los, M.J. Human induced pluripotent stem cell differentiation and direct transdifferentiation into corneal epithelial-like cells. Oncotarget 2016, 7, 42314–42329. [Google Scholar] [CrossRef]
  132. Richards, M.; Fong, C.-Y.; Tan, S.; Chan, W.-K.; Bongso, A. An Efficient and Safe Xeno-Free Cryopreservation Method for the Storage of Human Embryonic Stem Cells. Stem Cells 2004, 22, 779–789. [Google Scholar] [CrossRef] [PubMed]
  133. Kaur, J.; Tilkins, M.L.; Eckert, R.; Boucher, S. Methods for culturing human embryonic stem cells in a xeno-free system. Methods Mol. Biol. 2013, 997, 115–126. [Google Scholar] [CrossRef]
  134. Tanjore, H.; Kalluri, R. The role of type IV collagen and basement membranes in cancer progression and metastasis. Am. J. Pathol. 2006, 168, 715–717. [Google Scholar] [CrossRef] [Green Version]
  135. Kalluri, R. Basement membranes: Structure, assembly and role in tumour angiogenesis. Nat. Rev. Cancer 2003, 3, 422–433. [Google Scholar] [CrossRef] [PubMed]
  136. Cameron, J.D.; Skubitz, A.P.; Furcht, L.T. Type IV collagen and corneal epithelial adhesion and migration. Effects of type IV collagen fragments and synthetic peptides on rabbit corneal epithelial cell adhesion and migration in vitro. Investig. Ophthalmol. Vis. Sci. 1991, 32, 2766–2773. [Google Scholar] [PubMed]
  137. 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] [Green Version]
  138. 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] [CrossRef] [PubMed]
  139. Man, R.C.; Yong, T.K.; Hwei, N.M.; Halim, W.H.W.A.; Zahidin, A.Z.M.; Ramli, R.; Saim, A.B.; Idrus, R.B.H. Corneal regeneration by induced human buccal mucosa cultivated on an amniotic membrane following alkaline injury. Mol. Vis. 2017, 23, 810–822. [Google Scholar]
  140. Gaddipati, S.; Muralidhar, R.; Sangwan, V.S.; Mariappan, I.; Vemuganti, G.K.; Balasubramanian, D. Oral epithelial cells transplanted on to corneal surface tend to adapt to the ocular phenotype. Indian J. Ophthalmol. 2014, 62, 644–648. [Google Scholar] [CrossRef]
  141. Dobrowolski, D.; Orzechowska-Wylegala, B.; Wowra, B.; Wroblewska-Czajka, E.; Grolik, M.; Szczubialka, K.; Nowakowska, M.; Puzzolo, D.; Wylegala, E.A.; Micali, A.; et al. Cultivated Oral Mucosa Epithelium in Ocular Surface Reconstruction in Aniridia Patients. Biomed. Res. Int. 2015, 2015, 281870. [Google Scholar] [CrossRef] [Green Version]
  142. Chen, H.C.; Chen, H.L.; Lai, J.Y.; Chen, C.C.; Tsai, Y.J.; Kuo, M.T.; Chu, P.H.; Sun, C.C.; Chen, J.K.; Ma, D.H. Persistence of transplanted oral mucosal epithelial cells in human cornea. Investig. Ophthalmol. Vis. Sci. 2009, 50, 4660–4668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Reza, H.M.; Ng, B.-Y.; Gimeno, F.L.; Phan, T.T.; Ang, L.P.-K. Umbilical Cord Lining Stem Cells as a Novel and Promising Source for Ocular Surface Regeneration. Stem Cell Rev. Rep. 2011, 7, 935–947. [Google Scholar] [CrossRef]
  144. Ngan, N.D.; Chau, H.M.; Dong, P.N.; Cung, L.X.; Thuy, N.T.; Thang, P.T.; Thai, T.V.; Nga, V.T.; Bac, N.D. Tissue-Cultured Human Cord Lining Epithelial Cells in Treatment of Persistent Corneal Epithelial Defect. Open Access Maced. J. Med. Sci. 2019, 7, 4266–4271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Summary of signaling pathways involved in the differentiation of pluripotent cells into corneal epithelial cells. (a) Inhibition of Wnt secretion: IWP2 blocks porcupine, leading to reduction of palmitoylated Wnt protein so Wnt cannot interact with Wntless (WLS) which transports Wnt to plasma membrane; (b) Signaling pathways in regulating the corneal epithelial differentiation of pluripotent cells. Firstly, inhibition of Wnt pathway: (1) Blocking initiation of Wnt pathway by blocking Wnt (SFRP), LRP5/6 receptor (Dkks), and Frizzled receptor (RNF43, ZNRF3, SFRP); (2) Increasing degradation of β-catenin by IWR1 lead to block transcription of Wnt target genes; (3) Decreasing translocation of β-catenin from the cytoplasm into cell nucleus by RA, SU6656 leading to decrease in expression of Wnt target genes. Secondly, the activation FGF signaling pathway by supplement bFGF. Thirdly, the activation BMP signaling pathway by using BMP4. Lastly, inhibition TGF-β signaling by using inhibitors of TGF-β type I receptor (SB505124, SB431542, A83-01, A77-01).
Figure 1. Summary of signaling pathways involved in the differentiation of pluripotent cells into corneal epithelial cells. (a) Inhibition of Wnt secretion: IWP2 blocks porcupine, leading to reduction of palmitoylated Wnt protein so Wnt cannot interact with Wntless (WLS) which transports Wnt to plasma membrane; (b) Signaling pathways in regulating the corneal epithelial differentiation of pluripotent cells. Firstly, inhibition of Wnt pathway: (1) Blocking initiation of Wnt pathway by blocking Wnt (SFRP), LRP5/6 receptor (Dkks), and Frizzled receptor (RNF43, ZNRF3, SFRP); (2) Increasing degradation of β-catenin by IWR1 lead to block transcription of Wnt target genes; (3) Decreasing translocation of β-catenin from the cytoplasm into cell nucleus by RA, SU6656 leading to decrease in expression of Wnt target genes. Secondly, the activation FGF signaling pathway by supplement bFGF. Thirdly, the activation BMP signaling pathway by using BMP4. Lastly, inhibition TGF-β signaling by using inhibitors of TGF-β type I receptor (SB505124, SB431542, A83-01, A77-01).
Ijms 21 07834 g001
Table 1. Corneal epithelial differentiation of human MSCs in the co-culture system.
Table 1. Corneal epithelial differentiation of human MSCs in the co-culture system.
Human MSCsSignal Providing CellsCulture MediumDuration
(Days)
Marker Expression at the Final Differentiation StageReference
AT-MSCsporcine limbal epithelial stem cellsDMEM
FBS 10%
14gene expression
increase: CK3, CK12, p63
decrease: CD73, CD90, CD105, CK15, CK19, Cx43, ABCG2
[119]
CJ-MSCshuman corneal epithelial cellsSHEM medium
FBS 5%
DMSO 0.5%
EGF 5 ng/mL
Insulin 5 μg/mL
Transferrin 5 μg/mL
Sodium selenite
5 ng/mL
Hydrocortisone
0.5 μg/mL
Cholera toxin A
30 ng/mL
24gene expression
increase: CK3, CK8, CK12, CK14, CK15, ABCG2, DSC1, DSG1, NP63-α, Nestin, Involucrin
ICC
positive: CK3
[24]
EDT-MSCshuman corneal epithelial cellsDMEM/Ham F12 (1:1)
FBS 5%
DMSO 0.5%
EGF 5 ng/mL
21gene expression
increase: CK3, CK19
ICC
positive: CK3, CK19
[120]
Legend: DMEM, Dulbecco’s modified Eagle’s Medium; SHEM, supplemental hormonal epithelial medium; FBS, fetal bovine serum; DMSO, dimethyl sulfoxide.
Table 2. Corneal epithelial differentiation of human MSCs by defined induction media.
Table 2. Corneal epithelial differentiation of human MSCs by defined induction media.
Human MSCsInduction MediumDuration
(Days)
Marker Expression at the Final Differentiation StageReference
CJ-MSCsCnT-Prime 3D medium24gene expression
increase: CK3, CK8, CK12, CK14, CK15, ABCG2, DSC1, DSG1, NP63-α, Nestin, Involucrin
ICC
positive: CK3
[24]
CJ-MSCsDMEM: Ham’s F-12 (3:1)
FBS 5%
EGF 10 ng/mL
Insulin 5 μg/mL
Hydrocortisone 0.5 μg/mL
Triiodothyronine 2 nM
Adenine 2 nM
21gene expression
increase: CK3, CK8, CK12, DSC1, DSG1
[121]
BM-MSCsStep 1
DMEM
EGF 10 ng/mL
BMP4 25 ng/mL
All-trans retinoic acid 1 μM
Step 2
DMEM: Ham’s F-12 (3:1)
FBS 5%
EGF 10 ng/mL
Insulin 5 μg/mL
Hydrocortisone 0.5 μg/mL
Triiodothyronine 2 nM
Adenine 2 nM
4 (step 1) and
12 (step 2)
gene expression
increase: CK3, CK8, CK12, DSC1, DSG1
decrease: Oct4, Sox2, Nanog, Rex1, p63, ABCG2
ICC
positive: CK3, CK8, CK12, CK14, CK15, β-integrin, E-cadherin
negative: α-SMA
[20]
BM-MSCsDMEM
FBS 2%
L-glutamine 2 mM
All-trans retinoic acid 1 μM
7gene expression
increase: CK3, CK12, CK19, E-cad, ITGB1, Wnt-2, Snail
decrease: Vimentin
Protein expression
increase: CK12, CK19, ITGB1, N-cad
decrease: CK3, Vimentin, Snail, α-sma
[21]
BM-MSCsSHEM/Ham’s F12 (2:1)
FBS 2%
DMSO 0.5%
L-Glutamine 2 mM
EGF 10 ng/mL
Insulin 5 μg/mL
Hydrocortisone 0.4 μg/mL
Triiodothyronine 2 nM
Adenine 0.18 mM
7gene expression
increase: CK3, CK12, ITGB1
decrease: CK19, E-cad, Vimentin, Wnt-2, Snail
Protein expression
increase: CK3, CK12, ITGB1
decrease: CK19, Vimentin, Snail, α-sma, N-cad
[21]
Table 3. Corneal epithelial differentiation of human PSCs.
Table 3. Corneal epithelial differentiation of human PSCs.
Human Cell LineMethodStep 1Step 2Step 3Duration (Days)ICC ResultFlow Cytometry/Cytospin ResultGene Expression ResultReference
ESCs
(H1,
hES-NCL1)
CM of MMC-treated limbal fibroblasts3LG-DMEM/1F12
+hydrocortisone, insulin, adenine, EGF, tri-iodothyronine, cholera toxin
- 21hES-NCL1: high positive with CK12, CK3/12, p63 on D7Flow cytometry
p63 highest on D6
hES-NCL1: CK3/12~55% highest on D6
H1: CK3/12~55% highest on D9
CK3 highest on D15
hES-NCL1: CK3+, CK12+ highest on D6
H1: CK3+++, CK12+++ highest on D9
[25]
iPSCs
(L1B41, L1C51, 253G1, 201B7, L1B34)
Co-culture with MMC-treated PA6GMEM
+KSR, 2-mercaptoethanol
12–16 weeksL1B41: CK12, CK3, PAX6, CK14 L1B41 high responsive:
CK3, CK12: high express from week 8–12
Other cell lines low or non-responsive
[137]
ESCs
(H9, H3, H14)
iPSCs (6-9-9, 19-9-11)
Defined mediumDMEM/F12
+β-mercaptoethanol, DMSO, SU6656
DMEM/F12
+RA, BMP4,
bFGF, β-mercaptoethanol
DKSFM19CK14, CK3, p63Flow cytometry
CK3 5%, CK14 90.7%
[59]
iPSCsDefined mediumRegES medium
+SB505124, IWP-2, bFGF
Cnt-30 44D20
CK12, CK3, CK15, Ki67, Pax6
Cytospin on D44
p63 70%, CK3 30%,
CK15 55%, CK12 70%
P63+++, CK15++, CK3+, and CK12+[61]
ESCs
(H9,
RCM1)
CM from limbal fibroblasts3LG-DMEM/1F12
+hydrocortisone, insulin, adenine, EGF, triiodothyronine,
cholera toxin
21-Flow cytometry
H9: CK3 99% on D21, high rate from D4–21
RCM1: CK12 94% on D21, high rate from D16–21
CK3, CK12: very low express,
p63, CK19, ABCG2: high express on D16 and D21
[130]
iPSCsCM from limbal stromal cellsEpiLife medium
+human corneal growth supplement
21CK3, CK12, p63 express from D7–21 CK3, CK12 high express on D14–21[131]
iPSCs (UTA.045111.WT)Defined mediumXF-Ko-SR
D1: blebbistatin
D2: SB505124, bFGF
D3–4: BMP4
Cnt-30 21CK14, CK15, p63, PAX6Flow cytometry
p63 71%
[85]
ESCs (H9)
iPSCs
(SBAd2,
SBAd3)
Defined mediummTeSR1
D1:Y27632
DMEM/F12
+BMP4/RA/EGF/LDN193189
or BMP4+RA+EGF
or SB505124+IWP2 w/wo BMP4
CnT prime +10%FBS20CK12: not different on D20,
CK3: lower express on D20 in SBAd3 group
BMP4+RA+EGF is the best
SBAd2 &H9 responsive:
CK3, CK12: high express
SBAd3 non-responsive
[83]
ESCs
(H1, H9, CT3, Envy (GFP+))
Defined mediumE6
+IWR1, A83-01, bFGF w/wo BMP4
E6 75CK12, CK3Flow cytometry
CK3 55.6%, CK12 28.2%
[76]
ESCs
(Regea 08/017, Regea11/013)
iPSCs (UTA.04607.WT)
Defined mediumXF-Ko-SR
D1: blebbistatin
D2: SB505124, bFGF
D3–4: BMP4
Cnt-30 24D11: ABCG2+++, ∆Np63++, LGR5+++
D24: ABCG2+, ∆Np63+++, LGR5+, CK14+++, CK15++
Cytopsin:
D11: ABCG2 62.4%, ∆Np63 23.2%, CD200 42.6%
D24: ABCG2 1.8%, ∆Np63 54.3%, CK15 37%, CK14 56.2%
[87]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Theerakittayakorn, K.; Thi Nguyen, H.; Musika, J.; Kunkanjanawan, H.; Imsoonthornruksa, S.; Somredngan, S.; Ketudat-Cairns, M.; Parnpai, R. Differentiation Induction of Human Stem Cells for Corneal Epithelial Regeneration. Int. J. Mol. Sci. 2020, 21, 7834. https://doi.org/10.3390/ijms21217834

AMA Style

Theerakittayakorn K, Thi Nguyen H, Musika J, Kunkanjanawan H, Imsoonthornruksa S, Somredngan S, Ketudat-Cairns M, Parnpai R. Differentiation Induction of Human Stem Cells for Corneal Epithelial Regeneration. International Journal of Molecular Sciences. 2020; 21(21):7834. https://doi.org/10.3390/ijms21217834

Chicago/Turabian Style

Theerakittayakorn, Kasem, Hong Thi Nguyen, Jidapa Musika, Hataiwan Kunkanjanawan, Sumeth Imsoonthornruksa, Sirilak Somredngan, Mariena Ketudat-Cairns, and Rangsun Parnpai. 2020. "Differentiation Induction of Human Stem Cells for Corneal Epithelial Regeneration" International Journal of Molecular Sciences 21, no. 21: 7834. https://doi.org/10.3390/ijms21217834

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop