Int J Med Sci 2017; 14(8):705-710. doi:10.7150/ijms.19018 This issue Cite

Review

Characterization and Prospective of Human Corneal Endothelial Progenitors

Yongsong Liu1*, Hong Sun2*, Ping Guo3* Corresponding address, Min Hu4, Yuan Zhang5, Sean Tighe5, Shuangling Chen5, Yingting Zhu5 Corresponding address

1. Department of Ophthalmology, Yan' An Hospital of Kunming City, Kunming, 650051, China;
2. Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China;
3. Shenzhen Eye Hospital, School of Optometry & Ophthalmology of Shenzhen University, Shenzhen Key Laboratory of Department of Ophthalmology, Shenzhen, 518000, China;
4. Department of Ophthalmology, the Second People's Hospital of Yunnan Province, Kunming, 650021, China;
5. Research and Development Department, TissueTech, Inc., 7000 SW 97th Avenue, Suite 212, Miami, FL 33173, USA.
* The first three authors contributed equally to this manuscript.

Citation:
Liu Y, Sun H, Guo P, Hu M, Zhang Y, Tighe S, Chen S, Zhu Y. Characterization and Prospective of Human Corneal Endothelial Progenitors. Int J Med Sci 2017; 14(8):705-710. doi:10.7150/ijms.19018. https://www.medsci.org/v14p0705.htm
Other styles

File import instruction

Abstract

Corneal endothelial cells play a critical role in maintaining corneal transparency and dysfunction of these cells caused by aging, diseases (such as Fuch's dystrophy), injury or surgical trauma, which can lead to corneal edema and blindness. Due to their limited proliferative capacity in vivo, the only treatment method is via transplantation of a cadaver donor cornea. However, there is a severe global shortage of donor corneas. To circumvent such issues, tissue engineering of corneal tissue is a viable option thanks to the recent discoveries in this field. In this review, we summarize the recent advances in reprogramming adult human corneal endothelial cells into their progenitor status, the expansion methods and characteristics of human corneal endothelial progenitors, and their potential clinical applications as corneal endothelial cell grafts.

Keywords: Cornea, Endothelial, Progenitors, Tissue Engineering.

Introduction

The human corneal tissue is composed of different layers including a stratified epithelium, Bowman's layer, stroma, Descemet's membrane, and endothelium at the inner surface. The endothelial cells originate from cranial neural crest cells, forming a single monolayer of hexagonal cells lining the Descemet's membrane of the posterior cornea [1], and play a critical role in mediating vision function [2]. For example, corneal endothelium maintains the corneal transparency, stromal hydration and vision by mediating hydration (termed pump function) and preventing aqueous fluid from entering the stroma while also allowing permeability of nutrients (termed barrier function) (reviewed in [2, 3]). Unlike other species, human corneal endothelial cells (HCEC) are notorious for their limited proliferative capacity in vivo [4] due the mitotic block at the G1 phase in the cell cycle [5]. Hence if the endothelium were to become injured or become dysfunctional, there would be no proliferation to compensate for the cell loss and corneal blindness may occur [6, 7]. Until now, the only effective medical treatment is corneal transplantation from healthy donor cadavers. However due to the increasing aging population globally [8], there is an increasingly shortage of donor supply. Thus, it becomes necessary to seek alternative treatment options and one such promising therapeutic modality is the successful engineering of HCEC surgical grafts. In this review, we will discuss the current knowledge of adult corneal endothelial stem cells or progenitors with limited differentiation potential, the engineering of such HCEC grafts, and the potential application of HCEC tissue engineering.

Difference between ESC, iPSC and Adult Progenitors

Stem cells include embryonic stem cells (ESC), induced pluripotent stem cells (iPSC) and adult stem cells (progenitors). Although ESC have unlimited capacity for self-renewal and powerful pluripotency to differentiate into any type of cells in the human body theoretically, immune-rejection, teratoma formation, induction uncertainty and ethical concerns have hampered their progress towards any clinical applications. Since discovery of iPSC [9], a number of advantages have been proposed, such as a possible autologous approach to circumvent problems of immune-rejection and ethical concerns. Nevertheless, serious safety problems have been raised over the use of retroviral or lentiviral vectors in the creation of iPSC, which may induce genomic alteration and carcinogenesis [10-12]. In addition, their differentiation potential is uncertain. On the other hand, adult progenitors or somatic stem cells have limited differentiation potential, are located in a number of adult tissues such as the bone marrow, brain, heart, limbus, skeletal muscle and skin (reviewed in [13]) and their use avoids the ethical and differentiation potential concerns [14-17]. However, as such the case for all the aforementioned cells, progenitor stem cells are not easy to isolate, expand, and be maintained.

Origination of Human Corneal Endothelial Progenitors (HCEP)

Human corneal endothelial cells have been discovered and characterized over the last few decades [18-22]. In 1982, a group of so-called Schwalbe's line cells were first reported by Raviola et al [23]. These cells are located beneath the Schwalbe's ring, forming a discontinuous cord in the transition region of the anterior part of the trabecular meshwork and the corneal endothelium [23]. After laser trabeculoplasty, the proliferation of these cells was noted, suggesting that Schwalbe's line cells may have progenitor cell-like properties [23]. A similar observation was also reported in human laser-treated explants [24]. In another study, functional corneal endothelial cells were generated from these progenitors with a high proliferative potential and lineage [25].

It was found that HCEC from the corneal periphery and not the central area proliferated suggesting the presence of progenitor cells only in the peripheral area of the cornea (unpublished data). These results are consistent with the observance of telomerase activity in the peripheral and middle corneal areas, but not in the central cornea [26-28]. In the past decade, these results have been confirmed by a number of published articles that have suggested that endothelial progenitors are in fact present in the human cornea (reviewed in [29]). However, no specific markers have been used to identify endothelial progenitors in vivo. Therefore, the origin of the endothelial progenitors still cannot be clearly defined.

Expansion of HCEP

A scraping method was first used to isolate HCEC from the cornea and unlock their mitotic block ex vivo [30, 31]. Explant culture has been used for the expansion of HCEC up to 6 months and such expansion may produce small, hexagonal cells [32]. It has been found the sphere number were much higher in the peripheral area than that in the central area of the cornea, indicating a higher rate of self-renewal capability from the cells in the peripheral area [33]. Corneal endothelial aggregates (spheres) express a number of neural crest markers and may differentiate into various neuronal lineages [34], which is not surprising considering the corneal endothelium originates from neural crest cells in the embryonic development [25, 35]. HCEC in the spheres are small and hexagonal and are able to expand at a higher density with a higher number of BrdU-positive labeling, suggesting that HCEC sphere culture contain endothelial progenitors [34]. In a rabbit model, injection of corneal endothelial progenitor spheres into the eye restored the endothelial function and resulted in decreased corneal edema [36]. In addition, when cultured on denuded human amniotic membranes, these cells show a typical hexagonal shape and healthy tight junctions as determined by immunostaining of ZO-1 [37, 38].

Interestingly, when the cells are incubated in 0.02% EDTA for an hour, expression of neuronal markers is not observed even in the spheres (Reviewed in [39]). In fact, an EDTA/trypsin method has been developed to unlock the mitotic block of in vitro HCEC by dissociating their intercellular junctions and perturbing contact inhibition [20, 40], then culturing resultant single cells in bFGF- and serum-containing media [19, 20, 25, 40-45]. However, these conventional approaches can potentially trigger endothelial-mesenchymal transition (EMT), leading to the loss of the HCEC phenotype [7, 46], and loss of their progenitor status [47]. Such change of phenotype is due to activation of canonical Wnt signaling in the presence of EGF and/or bFGF, and even more-so when TGF-β1 is added, which activates canonical TGF-β signaling resulting in nuclear translocation of pSmad2/3 and Zeb1/2 [48]. Interestingly, the use of SB431542, a selective inhibitor of the TGF-β receptor, may block EMT in HCEC [49]. With this in mind, the blockade of canonical Wnt-Smad2/3-Zeb1/2 signaling is necessary during the expansion of HCEC.

In contrast to EDTA/trypsin, collagenase removes interstitial but not basement membrane of the corneal tissue [20]. Such resulting aggregates can be expanded effectively in a medium containing LIF and bFGF [35, 47]. LIF has been shown to delay contact-inhibition and is significantly more effective in promoting HCEP growth with bFGF [47, 50]. Many substrates for culturing these HCEP and HCEC [18, 22, 40, 41, 51], including artificial matrices, such as collagen I and fibronectin (FNC) [52], chondroitin sulfate and laminin [19], laminin-5 [53], matrigel [51] and FNC coating mix [27]. We have selected Collagen IV as the coating substrate because collagen IV has been identified as a better substrate for expansion of HCEP for tissue engineering purposes [20, 29, 54]. Although there has been reported successful amplification of HCEC [55, 56], up to now, no clinical application of cultured HCEC grafts has been reported.

Characterization of HCEP

A distinct subpopulation of cultured corneal endothelial cells has been discovered, showing colony-like structures with small size [57]. These cells are heterogeneous, have characteristic sphere growth tendency and plasticity to change to other type of cells, with high proliferative potential, dependent on endogenous upregulation of telomerase [25, 58] (also reviewed in [34]). The corneal endothelial progenitors are characterized as a group of small endothelial cells expressing p75NTR, SOX9, FOXC2, Twist, Snail and Slug with higher proliferative potential [25, 35, 59-63]. In addition, we have characterized human corneal endothelial progenitors as a group of cells expressing a number of ESC markers, such as cMyc, KLF4, Nanog, Nestin, Oct4, Rex1, Sox2, SSEA4 and NC markers such as AP2α, AP2β, FOXD3, HNK1, MSX1, p75NTR and Sox9 [47].

Reprogramming of HCEP as a Novel Strategy of Engineering HCEC

BMP signaling is necessary for programming of ESC to vascular endothelial cells [64, 65], and are important for reprogramming iPSC [66]. Recently, we have also reported that p120-RhoA-ROCK signaling may activate and elicit canonical BMP signaling in the growth of HCEC in MESCM [47] by weekly treatment of p120-Kaiso siRNAs for 5 weeks. Such expansion is associated with translocation of membranous p120 to the nucleus and release of nuclear Kaiso, a transcriptional repressor. That is, contact inhibition of HCEC monolayers can be safely perturbed by transient knockdown with p120 catenin (hereafter p120) ± Kaiso siRNAs to activate p120-Kaiso signaling via eliciting nuclear translocation of membranous p120 and nuclear release of the transcription repressor Kaiso. This then leads to RhoA-ROCK-canonical BMP signaling [47] when cultured in LIF-containing MESCM but non-canonical BMP-NFκB signaling when cultured in EGF-containing SHEM [67]. The former but not the latter also results in significant expansion of HCEC monolayers due to reprogramming into neural crest (NC) progenitors [47].

LIF, a member of the IL-6 family, is a key cytokine for sustaining self-renewal and pluripotency of mouse ESC and iPSC [68-71]). Upon binding to the LIF receptor, LIF activates JAK, which phosphorylates latent STAT3. pSTAT3 dimerizes and enters the nucleus to target expression of KLF4 [72] and Nanog [73]. We have recently reported that LIF-JAK1-STAT3 signaling indeed operates in HCEC monolayers cultured in MESCM [50]. The mechanism for such reprogramming is activation of the autoregulatory network of Oct4-Sox2-Nanog and miR-302 cluster in promoting self-renewal and pluripotency [67]. In this process, nucleus-translocated Oct4, Sox2, and Nanog directly binds to the promoter to activate expression of this miR-302 cluster [74, 75], and miR-302 then indirectly induces expression of Oct4, Sox2, and Nanog by reducing the expression of developmental genes [76, 77]. This approach is justified by our recent report showing that the reprogramming of NC progenitors also involves overexpression of miR 302b/c, which is completely blocked by RhoA inhibitor CT-04, ROCK1/2 siRNAs and BMP inhibitor Noggin [47], suggesting the overexpression of miR 302b/c is mediated by RhoA-ROCK1/2 signaling. This reprogramming resembles what has previously been reported [78, 79] that forced expression of transcription factors, e.g., Oct4, Sox2, KLF4 and c-Myc (SKOM), is a novel strategy [80] to reprogram somatic cells to iPSC [81, 82].

Barrier for Reprogramming HCEP

In mammalian cells, the G1/S transition is blocked in "contact inhibition" but facilitated in proliferation by E2F, of which the activity is inhibited by non-phosphorylated retinoblastoma tumor suppressor (Rb) [83]. Release of inhibition mediated by phosphorylation of Rb is controlled positively by cyclin D1/cyclin-dependent kinase-4 (CDK4) and cyclin E/CDK2 complex, but negatively by cyclin-dependent kinase inhibitors (CKIs) such as p16INK4a, p15INK4b, p18INK4c, p19INK4d, p21CIP1, p27KIP1, and p57KIP2 [84]. Without p120-Kaiso knockdown, we have recently reported that LIF-JAK1-STAT3 signaling delays contact inhibition [50]. MESCM without LIF, but not bFGF, delays contact inhibition by preventing nuclear translocation of p16INK4a, a process blocked by STAT3 siRNA [50].

Bmi-1, a member of the Polycomb Group (PcG) gene family of proteins that function as chromatin modifiers, is a suppressor of the Ink4a locus including p16INK4a [85-87]. p16INK4a belongs to the family of cyclin-dependent kinase inhibitors involved in cell cycle arrest at the G1 phase [88]. Nuclear p16INK4a is a hallmark of contact inhibition because p16INK4a binds to CDK4/6 inhibiting its kinase activity thereby preventing Rb phosphorylation during G1 to S transition [reviewed in [89]]. Hence p16INK4a controls HCEC senescence [77] and reprogramming [88, 90, 91]. p120-Kaiso knockdown releases nuclear Kaiso to the cytoplasm [46, 47], and activates both Rb and p16INK4a (reviewed in [92]), thus it is speculated that the mitotic block mediated by p16INK4a facilitates contact inhibition and senescence as a barrier against reprogramming and that such a barrier can be overcome by nuclear translocation of pBmi-1 facilitated by both STAT3 signaling and nuclear release of Kaiso. The aforementioned delay in contact inhibition may also be achieved by transit activation of LIF-JAK1-STAT3 signaling that also delays eventual nuclear translocation of p16INK4a [50]. Thus, JAK2-STAT3-Bmi-1 signaling is another downstream signaling of p120-Kaiso-RhoA-ROCK signaling that participates in reprogramming of HCEC into progenitors via inhibition of p16INK4a-mediated senescence [93].

Potential Clinical Application of Human Corneal Endothelial Grafts after Preclinical Animal Studies

Pre-clinical animal studies are the required method for examination of the safety and efficacy of human corneal endothelial grafts, including those expanded from HCEP. Because Descemet's stripping automated endothelial keratoplasty (DSAEK) and Descemet's membrane endothelial keratoplasty (DMEK) has become a standard procedure for corneal transplantation in patients with endothelial dysfunction in the last decades [94-97], transplanting only the HCEC sheets has become a standard procedure for treatment of CEC dysfunction [42]. In 2001, primary cultured HCEC were constructed onto the denuded Descemet's membrane for a test transplantation of ex vivo human corneal endothelium [40]. In this case, the recipient cornea was cultivated in organ culture for up to 2 weeks. The mean endothelial cell density in the transplanted corneas was 1895 cells/mm2 (1503-2159 cells/mm2), and was deemed a success [40]. Amniotic membrane has also been introduced as a reliable carrier for cultured HCEC transplantation [41]. The density of the HCEC on the amniotic membrane was shown to be greater than 3000 cells/mm2, similar to that of in vivo density. Another potential carrier is collagen I which has been successfully used for cultured monkey corneal endothelial sheets [98, 99] and we are currently testing in a mini-pig model with cultured endothelial grafts.

The current limitations and challenges for the research of HCEP are there are many difficulties for isolation and expansion of a population of HCEP without contamination of other type of cells and without change of the cell phenotype as in vitro culture time passes by. Therefore, there is no cell-based therapies for cure of human corneal endothelial dysfunction so far. However, the research in this field has progressed rapidly. Hopefully, we will resolve those issues in the near future.

Conclusions

This review has highlighted the latest discoveries and innovations in corneal endothelial engineering. The novel techniques presented here demonstrate the potential future treatments of CEC dysfunction.

Abbreviations

bFGF: Basic fibroblast growth factor; Bmi-1: B lymphoma Mo-MLV insertion region 1 homolog; BMP: Bone morphogenic protein; BrdU: Bromodeoxyuridine; EDTA: Ethylenediaminetetraacetic acid; DMEK: Descemet's membrane endothelial keratoplasty; DSAEK: Descemet's stripping automated endothelial keratoplasty; EMT: Endothelial-mesenchymal transition; EGF: Epidermal growth factor; ESC: embryonic stem cell; FNC: fibronectin; HCEC: human corneal endothelial cell; HCEP: human corneal endothelial progenitor; IL: interleukin; iPSC: induced pluripotent stem cell; JAK: Janus kinase; KLF4: Kruppel-like factor 4; LIF: Leukemia inhibitory factor; MESCM: modified embryonic stem cell medium; NC: neural crest; NFkB: nuclear factor kappa-light-chain-enhancer of activated B cells; NGF: neural growth factor; p16INK4a: a tumor suppressor protein functions as an inhibitor of CDK4 and CDK6, the D-type cyclin-dependent kinases that initiate the phosphorylation of the retinoblastoma tumor suppressor protein; p120: p120 catenin; PcG: polycomb group; Rb: retinoblastoma; PKP: penetrating keratoplasty; Rho: Ras homolog gene family; ROCK: Rho-associated protein kinase; siRNA: Small interfering ribonucleic acid; SHEM: supplemental hormonal epithelial medium; STAT: signal transducer and activator of transcription; TGF: Transforming growth factor; ZO-1: Zona occludens protein 1

Acknowledgements

The work from this article is supported by Grant JCY20130401152829824 from Shenzhen Municipal Science and Technology Innovation Committee and Grant 2015A030313774 from Natural Science Fund of Guangdong Province, and R43 EY022502-01 and R44 EY022502-02 grants from the National Eye Institute, National Institutes of Health, Bethesda, MD, USA.

Authors Contributions

Yongsong Liu, Hong Sun, Ping Guo, Min Hu, Yuan Zhang, Sean Tighe and Shuangling Chen contributed to collection of information, organization and part of writings. Ping Guo and Yingting Zhu oversaw this project and finalized this review.

Competing Interests

The authors have declared that no competing interest exists.

References

1. Bahn CF, Falls HF, Varley GA, Meyer RF, Edelhauser HF, Bourne WM. Classification of corneal endothelial disorders based on neural crest origin. Ophthalmology. 1984;91:558-63

2. Bonanno JA. Identity and regulation of ion transport mechanisms in the corneal endothelium. Prog Retin Eye Res. 2003;22:69-94

3. Fischbarg J, Maurice DM. An update on corneal hydration control. Exp Eye Res. 2004;78:537-41

4. Laing RA, Neubauer L, Oak SS, Kayne HL, Leibowitz HM. Evidence for mitosis in the adult corneal endothelium. Ophthalmology. 1984;91:1129-34

5. Joyce NC. Cell cycle status in human corneal endothelium. Exp Eye Res. 2005;81:629-38

6. Bourne WM, McLaren JW. Clinical responses of the corneal endothelium. Exp Eye Res. 2004;78:561-72

7. Lee JG, Kay EP. FGF-2-mediated signal transduction during endothelial mesenchymal transformation in corneal endothelial cells. Exp Eye Res. 2006;83:1309-16

8. World Health Organization. Visual impairment and blindness. 2014.

9. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663-76

10. Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K. Induced pluripotent stem cells generated without viral integration. Science. 2008;322:945-9

11. Zhou H, Wu S, Joo JY, Zhu S, Han DW, Lin T. et al. Generation of induced pluripotent stem cells using recombinant proteins. Cell Stem Cell. 2009;4:381-4

12. Lin T, Ambasudhan R, Yuan X, Li W, Hilcove S, Abujarour R. et al. A chemical platform for improved induction of human iPSCs. Nat Methods. 2009;6:805-8

13. Mimeault M, Batra SK. Recent progress on tissue-resident adult stem cell biology and their therapeutic implications. Stem Cell Rev. 2008;4:27-49

14. Mimeault M, Hauke R, Batra SK. Stem cells: a revolution in therapeutics-recent advances in stem cell biology and their therapeutic applications in regenerative medicine and cancer therapies. Clin Pharmacol Ther. 2007;82:252-64

15. Lodi D, Iannitti T, Palmieri B. Stem cells in clinical practice: applications and warnings. J Exp Clin Cancer Res. 2011;30:9

16. MacLaren RE, Pearson RA, MacNeil A, Douglas RH, Salt TE, Akimoto M. et al. Retinal repair by transplantation of photoreceptor precursors. Nature. 2006;444:203-7

17. Limb GA, Daniels JT. Ocular regeneration by stem cells: present status and future prospects. Br Med Bull. 2008;85:47-61

18. Yue BY, Sugar J, Gilboy JE, Elvart JL. Growth of human corneal endothelial cells in culture. Invest Ophthalmol Vis Sci. 1989;30:248-53

19. Engelmann K, Bohnke M, Friedl P. Isolation and long-term cultivation of human corneal endothelial cells. Invest Ophthalmol Vis Sci. 1988;29:1656-62

20. Li W, Sabater AL, Chen YT, Hayashida Y, Chen SY, He H. et al. A novel method of isolation, preservation, and expansion of human corneal endothelial cells. Invest Ophthalmol Vis Sci. 2007;48:614-20

21. Peh GS, Toh KP, Wu FY, Tan DT, Mehta JS. Cultivation of human corneal endothelial cells isolated from paired donor corneas. PLoS One. 2011;6:e28310

22. Engelmann K, Friedl P. Optimization of culture conditions for human corneal endothelial cells. In Vitro Cell Dev Biol. 1989;25:1065-72

23. Raviola G. Schwalbe line's cells: a new cell type in the trabecular meshwork of Macaca mulatta. Invest Ophthalmol Vis Sci. 1982;22:45-56

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

25. Yokoo S, Yamagami S, Yanagi Y, Uchida S, Mimura T, Usui T. et al. Human corneal endothelial cell precursors isolated by sphere-forming assay. Invest Ophthalmol Vis Sci. 2005;46:1626-31

26. Wilson SE, Lloyd SA, He YG, McCash CS. Extended life of human corneal endothelial cells transfected with the SV40 large T antigen. Invest Ophthalmol Vis Sci. 1993;34:2112-23

27. Zhu C, Joyce NC. Proliferative response of corneal endothelial cells from young and older donors. Invest Ophthalmol Vis Sci. 2004;45:1743-51

28. Joyce NC, Harris DL, Mello DM. Mechanisms of mitotic inhibition in corneal endothelium: contact inhibition and TGF-beta2. Invest Ophthalmol Vis Sci. 2002;43:2152-9

29. Zhu YT, Tighe S, Chen SL, John T, Kao WY, Tseng SC. Engineering of Human Corneal Endothelial Grafts. Curr Ophthalmol Rep. 2015;3:207-17

30. Pistsov MY, Sadovnikova E, Danilov SM. Human corneal endothelial cells: isolation, characterization and long-term cultivation. Exp Eye Res. 1988;47:403-14

31. Gospodarowicz D, Mescher AL, Birdwell CR. Stimulation of corneal endothelial cell proliferations in vitro by fibroblast and epidermal growth factors. Exp Eye Res. 1977;25:75-89

32. Walshe J, Harkin DG. Serial explant culture provides novel insights into the potential location and phenotype of corneal endothelial progenitor cells. Exp Eye Res. 2014;127:9-13

33. Mimura T, Yamagami S, Yokoo S, Araie M, Amano S. Comparison of rabbit corneal endothelial cell precursors in the central and peripheral cornea. Invest Ophthalmol Vis Sci. 2005;46:3645-8

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

35. Hara S, Hayashi R, Soma T, Kageyama T, Duncan T, Tsujikawa M. et al. Identification and potential application of human corneal endothelial progenitor cells. Stem Cells Dev. 2014;23:2190-201

36. Mimura T, Yokoo S, Araie M, Amano S, Yamagami S. Treatment of rabbit bullous keratopathy with precursors derived from cultured human corneal endothelium. Invest Ophthalmol Vis Sci. 2005;46:3637-44

37. Yamagami S, Mimura T, Yokoo S, Takato T, Amano S. Isolation of human corneal endothelial cell precursors and construction of cell sheets by precursors. Cornea. 2006;25:S90-2

38. Mimura T, Yamagami S, Yokoo S, Usui T, Amano S. Selective isolation of young cells from human corneal endothelium by the sphere-forming assay. Tissue Eng Part C Methods. 2010;16:803-12

39. Parekh M, Graceffa V, Bertolin M, Elbadawy H, Salvalaio G, Ruzza A. Reconstruction and regeneration of the corneal endothelium: a review on the current methods and future aspects. Journal of Cell Science & Therapy. 2013;4:article146

40. Chen KH, Azar D, Joyce NC. Transplantation of adult human corneal endothelium ex vivo: a morphologic study. Cornea. 2001;20:731-7

41. Ishino Y, Sano Y, Nakamura T, Connon CJ, Rigby H, Fullwood NJ. et al. Amniotic membrane as a carrier for cultivated human corneal endothelial cell transplantation. Invest Ophthalmol Vis Sci. 2004;45:800-6

42. Mimura T, Yamagami S, Yokoo S, Usui T, Tanaka K, Hattori S. et al. Cultured human corneal endothelial cell transplantation with a collagen sheet in a rabbit model. Invest Ophthalmol Vis Sci. 2004;45:2992-7

43. Hsiue GH, Lai JY, Chen KH, Hsu WM. A novel strategy for corneal endothelial reconstruction with a bioengineered cell sheet. Transplantation. 2006;81:473-6

44. Sumide T, Nishida K, Yamato M, Ide T, Hayashida Y, Watanabe K. et al. Functional human corneal endothelial cell sheets harvested from temperature-responsive culture surfaces. FASEB J. 2006;20:392-4

45. Hatou S, Yoshida S, Higa K, Miyashita H, Inagaki E, Okano H. et al. Functional corneal endothelium derived from corneal stroma stem cells of neural crest origin by retinoic acid and Wnt/beta-catenin signaling. Stem Cells Dev. 2013;22:828-39

46. Zhu YT, Chen HC, Chen SY, Tseng SC. Nuclear p120 catenin unlocks mitotic block of contact-inhibited human corneal endothelial monolayers without disrupting adherent junctions. J Cell Sci. 2012;125:3636-48

47. Zhu YT, Li F, Han B, Tighe S, Zhang S, Chen SY. et al. Activation of RhoA-ROCK-BMP signaling reprograms adult human corneal endothelial cells. J Cell Biol. 2014;206:799-811

48. Chen HC, Zhu YT, Chen SY, Tseng SC. Wnt signaling induces epithelial-mesenchymal transition with proliferation in ARPE-19 cells upon loss of contact inhibition. Lab Invest. 2012;92:676-87

49. Okumura N, Kay EP, Nakahara M, Hamuro J, Kinoshita S, Koizumi N. Inhibition of TGF-beta signaling enables human corneal endothelial cell expansion in vitro for use in regenerative medicine. PLoS One. 2013;8:e58000

50. Liu X, Tseng SC, Zhang MC, Chen SY, Tighe S, Lu WJ. et al. LIF-JAK1-STAT3 signaling delays contact inhibition of human corneal endothelial cells. Cell Cycle. 2015;14:1197-206

51. Miyata K, Drake J, Osakabe Y, Hosokawa Y, Hwang D, Soya K. et al. Effect of donor age on morphologic variation of cultured human corneal endothelial cells. Cornea. 2001;20:59-63

52. Joyce NC, Zhu CC. Human corneal endothelial cell proliferation: potential for use in regenerative medicine. Cornea. 2004;23:S8-S19

53. Yamaguchi M, Ebihara N, Shima N, Kimoto M, Funaki T, Yokoo S. et al. Adhesion, migration, and proliferation of cultured human corneal endothelial cells by laminin-5. Invest Ophthalmol Vis Sci. 2011;52:679-84

54. Zhu YT, Hayashida Y, Kheirkhah A, He H, Chen SY, Tseng SC. Characterization and comparison of intercellular adherent junctions expressed by human corneal endothelial cells in vivo and in vitro. Invest Ophthalmol Vis Sci. 2008;49:3879-86

55. Engler C, Kelliher C, Wahlin KJ, Speck CL, Jun AS. Comparison of non-viral methods to genetically modify and enrich populations of primary human corneal endothelial cells. Mol Vis. 2009;15:629-37

56. Baum JL, Niedra R, Davis C, Yue BY. Mass culture of human corneal endothelial cells. Arch Ophthalmol. 1979;97:1136-40

57. Schmedt T, Chen Y, Nguyen TT, Li S, Bonanno JA, Jurkunas UV. Telomerase immortalization of human corneal endothelial cells yields functional hexagonal monolayers. PLoS One. 2012;7:e51427

58. Yoo H, Feng X, Day RD. Adolescents' empathy and prosocial behavior in the family context: a longitudinal study. J Youth Adolesc. 2013;42:1858-72

59. He Z, Campolmi N, Gain P, Ha Thi BM, Dumollard JM, Duband S. et al. Revisited microanatomy of the corneal endothelial periphery: new evidence for continuous centripetal migration of endothelial cells in humans. Stem Cells. 2012;30:2523-34

60. McGowan SL, Edelhauser HF, Pfister RR, Whikehart DR. Stem cell markers in the human posterior limbus and corneal endothelium of unwounded and wounded corneas. Mol Vis. 2007;13:1984-2000

61. Whikehart DR, Parikh CH, Vaughn AV, Mishler K, Edelhauser HF. Evidence suggesting the existence of stem cells for the human corneal endothelium. Mol Vis. 2005;11:816-24

62. Yamagami S, Yokoo S, Mimura T, Takato T, Araie M, Amano S. Distribution of precursors in human corneal stromal cells and endothelial cells. Ophthalmology. 2007;114:433-9

63. Yoshida S, Shimmura S, Nagoshi N, Fukuda K, Matsuzaki Y, Okano H. et al. Isolation of multipotent neural crest-derived stem cells from the adult mouse cornea. Stem Cells. 2006;24:2714-22

64. Suzuki Y, Montagne K, Nishihara A, Watabe T, Miyazono K. BMPs promote proliferation and migration of endothelial cells via stimulation of VEGF-A/VEGFR2 and angiopoietin-1/Tie2 signalling. J Biochem. 2008;143:199-206

65. Suzuki Y, Ohga N, Morishita Y, Hida K, Miyazono K, Watabe T. BMP-9 induces proliferation of multiple types of endothelial cells in vitro and in vivo. J Cell Sci. 2010;123:1684-92

66. Samavarchi-Tehrani P, Golipour A, David L, Sung HK, Beyer TA, Datti A. et al. Functional genomics reveals a BMP-driven mesenchymal-to-epithelial transition in the initiation of somatic cell reprogramming. Cell Stem Cell. 2010;7:64-77

67. Zhu YT, Han B, Li F, Chen SY, Tighe S, Zhang S. et al. Knockdown of both p120 catenin and Kaiso promotes expansion of human corneal endothelial monolayers via RhoA-ROCK-noncanonical BMP-NFkappaB pathway. Invest Ophthalmol Vis Sci. 2014;55:1509-18

68. Yang J, van Oosten AL, Theunissen TW, Guo G, Silva JC, Smith A. Stat3 activation is limiting for reprogramming to ground state pluripotency. Cell Stem Cell. 2010;7:319-28

69. Hirai H, Karian P, Kikyo N. Regulation of embryonic stem cell self-renewal and pluripotency by leukaemia inhibitory factor. Biochem J. 2011;438:11-23

70. van Oosten AL, Costa Y, Smith A, Silva JC. JAK/STAT3 signalling is sufficient and dominant over antagonistic cues for the establishment of naive pluripotency. Nat Commun. 2012;3:817

71. Mathieu ME, Saucourt C, Mournetas V, Gauthereau X, Theze N, Praloran V. et al. LIF-dependent signaling: new pieces in the Lego. Stem Cell Rev. 2012;8:1-15

72. Jiang J, Chan YS, Loh YH, Cai J, Tong GQ, Lim CA. et al. A core Klf circuitry regulates self-renewal of embryonic stem cells. Nat Cell Biol. 2008;10:353-60

73. Theunissen TW, van Oosten AL, Castelo-Branco G, Hall J, Smith A, Silva JC. Nanog overcomes reprogramming barriers and induces pluripotency in minimal conditions. Curr Biol. 2011;21:65-71

74. Huang Y, Sheha H, Tseng SC. Conjunctivochalasis interferes with tear flow from fornix to tear meniscus. Ophthalmology. 2013;120:1681-7

75. Tjiu JW, Chen JS, Shun CT, Lin SJ, Liao YH, Chu CY. et al. Tumor-associated macrophage-induced invasion and angiogenesis of human basal cell carcinoma cells by cyclooxygenase-2 induction. J Invest Dermatol. 2009;129:1016-25

76. Lin SL, Chang DC, Lin CH, Ying SY, Leu D, Wu DT. Regulation of somatic cell reprogramming through inducible mir-302 expression. Nucleic Acids Res. 2011;39:1054-65

77. Anokye-Danso F, Trivedi CM, Juhr D, Gupta M, Cui Z, Tian Y. et al. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell. 2011;8:376-88

78. Barroso-delJesus A, Lucena-Aguilar G, Sanchez L, Ligero G, Gutierrez-Aranda I, Menendez P. The Nodal inhibitor Lefty is negatively modulated by the microRNA miR-302 in human embryonic stem cells. FASEB J. 2011;25:1497-508

79. Card DA, Hebbar PB, Li L, Trotter KW, Komatsu Y, Mishina Y. et al. Oct4/Sox2-regulated miR-302 targets cyclin D1 in human embryonic stem cells. Mol Cell Biol. 2008;28:6426-38

80. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861-72

81. Drews K, Jozefczuk J, Prigione A, Adjaye J. Human induced pluripotent stem cells-from mechanisms to clinical applications. J Mol Med (Berl). 2012;90:735-45

82. Iglesias-Garcia O, Pelacho B, Prosper F. Induced pluripotent stem cells as a new strategy for cardiac regeneration and disease modeling. J Mol Cell Cardiol. 2013;62:43-50

83. DeGregori J, Leone G, Miron A, Jakoi L, Nevins JR. Distinct roles for E2F proteins in cell growth control and apoptosis. Proc Natl Acad Sci U S A. 1997;94:7245-50

84. Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev. 1995;9:1149-63

85. Jacobs JJ, Kieboom K, Marino S, DePinho RA, van Lohuizen M. The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus. Nature. 1999;397:164-8

86. Park IK, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL. et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature. 2003;423:302-5

87. Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF, Morrison SJ. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature. 2003;425:962-7

88. Banito A, Rashid ST, Acosta JC, Li S, Pereira CF, Geti I. et al. Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev. 2009;23:2134-9

89. Rayess H, Wang MB, Srivatsan ES. Cellular senescence and tumor suppressor gene p16. Int J Cancer. 2012;130:1715-25

90. Li H, Collado M, Villasante A, Strati K, Ortega S, Canamero M. et al. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature. 2009;460:1136-9

91. Hirosue A, Ishihara K, Tokunaga K, Watanabe T, Saitoh N, Nakamoto M. et al. Quantitative assessment of higher-order chromatin structure of the INK4/ARF locus in human senescent cells. Aging Cell. 2012;11:553-6

92. Carnahan RH, Rokas A, Gaucher EA, Reynolds AB. The molecular evolution of the p120-catenin subfamily and its functional associations. PLoS One. 2010;5:e15747

93. Lu WJ, Tseng SC, Chen S, Tighe S, Zhang Y, Liu X. et al. Senescence Mediated by p16INK4a Impedes Reprogramming of Human Corneal Endothelial Cells into Neural Crest Progenitors. Sci Rep. 2016;6:35166

94. Gorovoy MS. Descemet-stripping automated endothelial keratoplasty. Cornea. 2006;25:886-9

95. Koenig SB, Covert DJ. Early results of small-incision Descemet's stripping and automated endothelial keratoplasty. Ophthalmology. 2007;114:221-6

96. Terry MA, Shamie N, Chen ES, Hoar KL, Friend DJ. Endothelial keratoplasty a simplified technique to minimize graft dislocation, iatrogenic graft failure, and pupillary block. Ophthalmology. 2008;115:1179-86

97. Price MO, Baig KM, Brubaker JW, Price FW Jr. Randomized, prospective comparison of precut vs surgeon-dissected grafts for descemet stripping automated endothelial keratoplasty. Am J Ophthalmol. 2008;146:36-41

98. Koizumi N, Sakamoto Y, Okumura N, Okahara N, Tsuchiya H, Torii R. et al. Cultivated corneal endothelial cell sheet transplantation in a primate model. Invest Ophthalmol Vis Sci. 2007;48:4519-26

99. Koizumi N, Sakamoto Y, Okumura N, Tsuchiya H, Torii R, Cooper LJ. et al. Cultivated corneal endothelial transplantation in a primate: possible future clinical application in corneal endothelial regenerative medicine. Cornea. 2008;27(Suppl 1):S48-55

Author contact

Corresponding address Corresponding authors: Ping Guo: Shenzhen Eye Hospital, Zetian Road 18, Room 421, Futian District, Shenzhen, 518000, China. Tel 08613924659029; Fax 08675523959500; Email: 2607212858com; or Yingting Zhu, Ph.D. Research and Development Department, TissueTech, Inc., 7000 SW 97th Avenue, Suite 212, Miami, FL 33173. Telephone: (786) 456-7632; Fax: (305) 274-1297; E-mail: yzhucom


Received 2017-1-2
Accepted 2017-4-21
Published 2017-6-30


Citation styles

APA
Liu, Y., Sun, H., Guo, P., Hu, M., Zhang, Y., Tighe, S., Chen, S., Zhu, Y. (2017). Characterization and Prospective of Human Corneal Endothelial Progenitors. International Journal of Medical Sciences, 14(8), 705-710. https://doi.org/10.7150/ijms.19018.

ACS
Liu, Y.; Sun, H.; Guo, P.; Hu, M.; Zhang, Y.; Tighe, S.; Chen, S.; Zhu, Y. Characterization and Prospective of Human Corneal Endothelial Progenitors. Int. J. Med. Sci. 2017, 14 (8), 705-710. DOI: 10.7150/ijms.19018.

NLM
Liu Y, Sun H, Guo P, Hu M, Zhang Y, Tighe S, Chen S, Zhu Y. Characterization and Prospective of Human Corneal Endothelial Progenitors. Int J Med Sci 2017; 14(8):705-710. doi:10.7150/ijms.19018. https://www.medsci.org/v14p0705.htm

CSE
Liu Y, Sun H, Guo P, Hu M, Zhang Y, Tighe S, Chen S, Zhu Y. 2017. Characterization and Prospective of Human Corneal Endothelial Progenitors. Int J Med Sci. 14(8):705-710.

This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
Popup Image