ANNEX PUBLISHERS

Journal of Stem Cells and Clinical Practice

Open Access
Research Article
Max Screen

Introduction of Stem Cells in Ophthalmology

Copyright: © 2018 Soleimani M. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Related article at Pubmed, Google Scholar

Abstract

Purpose:In this article, we review main clinical trials reported in the past decade.

Methods:Stem cells are relatively undifferentiated, with unlimited proliferative ability; self-renewal capability and also they can differentiate into specialized cells. Somatic stem cells in adult organisms are responsible for regenerating and self-renewing tissue. Many different stem cell types reside in the eye.

Results:One of the most important stem cells is limbal stem cell that retains in an undifferentiated state and exists in an optimal microenvironment or “niche”. The importance of limbal stem cells in maintaining the corneal epithelium throughout life has long been recognized. Furthermore, stem cells in the retina have been suggested for treatment of the retinal degeneration, which generally results in constant visual disturbance or even blindness.

Conclusion:This review will briefly focus on the principal stem cells in the eye especially limbal stem cells.

Keywords: Limbal stem cell; Limbal stem cell deficiency; Transplantation; Retinal progenitor; Clinical trials

Introduction

Stem cells are undifferentiated cells with unlimited proliferative ability of self-renewal differentiating into differentiated cells. Limbal stem cells are nested in an optimal microenvironment or “niche”. The importance of limbal stem cells in maintaining the corneal epithelium throughout life has long been recognized. Limbal epithelium consists of a non-keratinized stratified squamous epithelium and located between corneal and conjunctival epithelia [1-5]. It has been found to include a source of stem cells (SCs) known as corneal epithelial SCs or limbal stem cells (LSCs). Limbal Stem Cell Deficiency (LSCD) is a heterogeneous group of diseases in which the limbal epithelial stem cells (LESCs) are depleted by excessive damage or disease in the limbus to replenish the consistent corneal epithelial regeneration [6]. There are several procedures to address LSCD; some of the most global procedures are keratolimbal allograft (KLAL), conjunctival limbal autograft (CLAU) and simple limbal epithelial transplantation (SLET). In this review, we want to focus on stem cell field in the ophthalmology especially limbal stem cells.

Methods

We conducted a review of the literature using PubMed database to gather relevant English articles with the keywords “stem cell” and “eye” or “ophthalmology”. Relevant articles during the past decade were selected.

Results
Limbal stem cells

Corneal epithelial stem cells:The ocular surface of the eye is covered with corneal epithelial surface, limbus, and conjunctiva [1]. Cornea has several roles such as: refraction, photo protection, transparency, and protection of internal ocular structures from the external environment [2,3]. Mature corneal epithelium is five or six layers of non-keratinizing stratified squamous epithelial cells with about 0.05 mm thickness and derived from the head surface ectoderm overlying the lens after invagination. The development of the cornea is a terminal inductive event in eye formation [4]. Conjunctival epithelium is the significant epithelium of the ocular surface expanding from the posterior margin of the eyelids, posterior surface to the peripheral edge of the tarsal plate, then folds back to the sclera and then continues as the limbal epithelium [1-5]. Limbal epithelium consists of a non-keratinized stratified squamous epithelium and located between corneal and conjunctival epithelia [6]. It has been found to include a source of SCs known as corneal epithelial SCs or LSCs [7,8].

Concept of LSC:The repair and regeneration throughout the life of the adult cornea is responsible by LSC population [9]. In order to replenish the SC population, only one of the daughter cells is divided asymmetrically and can re-enter the niche and become a stem cell [6]. The characteristics of these stem cells are undifferentiated, slow-cycling, self-renewal, to have high proliferative potential, small size, and high nuclear to cytoplasm ratio [10-12]. The other cell is destined for differentiation to transient amplifying cell (TAC) which migrates to the corneal epithelium divides at an exponential rate and will finally differentiate into a post-mitotic cell (PMC) that can no longer multiply. The PMCs differentiate and mature into terminally differentiated cells (TDC) that represent the final phenotypic expression of the tissue type [6].

LSC niche:Stem cells in all renewable tissues are normally located in a unique and appropriate microenvironment called niche, which supports self-renewal and multipotential activity and nurtures the stem cells. SCs organized in a ridge-like structure around the circumference of the cornea, which have been suggested the ridge to be the rudimentary niche for LSC [13]. Some studies indicate that the adult LSC niche exists within the basal interpalisade epithelial papillae of the Palisades of Vogt which contains radially-oriented fibrovascular ridges. These are concentrated along the superior and inferior limbus and found at the corneoscleral limbus [14]. The function of normal SC depends more on their niche than their gene expression patterns. Interaction between stem cells and their niches are critical for regulating SC function such as quiescence, apoptosis, division, or differentiation of stem cells [15,16]. SC behavior is regulated by wide variety of cells, including neighboring cells, signaling molecules (integrins, Wnt/βcatenin,and Notch), local environmental factors such as extracellular matrix, and other intercellular contacts [17-19]. The precise molecular mechanism by which the stromal niche regulates limbal stem cells is just beginning to be understood [20].

Limbal epithelial stem cell markers:The ability to identify SCs in different organs has been prevented by nonspecific and unreliable markers. α-enolase is expressed in embryonic basal cells and localize to limbal basal epithelial cells without cytokeratin 3 (CK 3) and it is suggested as a potential stem cell marker [21,22]. The relation of LESC [23] can identify the existence of associated markers (e.g. ABCG2, vimentin, and cytokeratin 19) and the lack expression of differentiation markers (e.g. CK 3/12, connexin 43, and involucrin). Many of these markers are identified in early TACs. Consequently, small size of cell, high nucleus to cytoplasm ratio and other morphological, phenotypic and functional characteristics of stem cells are used with stem cell markers [24]. In Table 1, some important markers are described [25-34].

Limbal stem cell deficiency: etiology and classification

World Health Organization (WHO) estimates that 10 million of 45 million bilaterally blind people worldwide are the result of corneal involvement [35]. LSCD is a heterogeneous group of diseases in which the LESCs are depleted by excessive damage or disease in the limbus to replenish the consistent corneal epithelial regeneration. LSCD occurs in genetic or acquired disorders. There are hereditary or genetic causes, such as aniridia, keratitis associated with multiple endocrine deficiencies, epidermal dysplasia (ectrodactyly-ectodermal dysplasia-clefting syndrome, Keratitis-ichthyosis-deafness (KID) Syndrome) [36,37]. Aniridia(developmental dysgenesis of the anterior segment of the eye) caused by mutations in the pax6 gene. Pax6 is essential for eye development (oculogenesis) and maintenance of LSC function.

Most commonly, LESC deficiency is often caused by acquired factors. There are acquired causes, including ionizing and ultraviolet radiation, extensive microbial infection, contact lens (CL) wear, industrial accidents, after multiple resections of ocular surface tumors and Johnson syndrome (SJS) and Ocular cicatricial pemphigoid (OCP) that appears through dysfunction in mucous membrane [38-40]. In other term, LSCD divided into primary or secondary. The primary, which does not present any identifiable external factors, is supported by an insufficient microenvironment. Here, there are some dysfunction/poor regulation of stromal microenvironment of limbal epithelial stem cells: congenital erythrokeratoderma, keratitis with multiple endocrine deficiency and poor nutritional or cytokine supply, neurotrophic keratopathy, peripheral inflammation and sclerocornea [41,42]. Secondary LSCD is acquired by external factors. Some causes of Secondary LSCD have also been described in acquired LSCD [40-42].

Clinical features of LSCD:Conjunctivalization of the cornea associated with goblet cells, superficial and deep vascularization, chronic inflammation, persistent epithelial defects and scarring are the essential clinical signs of LSCD [43-45]. The most important clinical trait of LSCD is conjunctivalization of the cornea that results in depletion of LESCs or loss of function by trauma or disease [46-48]. Tearing, reduced vision, chronic discomfort, chronic inflammation, stromal scarring, blepharospasm, photophobia, neovascularization, and persistent epithelial defects (PEDs) are clinical features of conjunctivalization [2,46]. It depends on the size of limbal damage, which has a pattern of partial ingrowths (partial LSCD) or will affect the whole cornea (total LSCD) [49]. Also, a variance in the thickness and transparency of the corneal epithelium on slit lamp examination may be seen [50]. The cause of the LSCD often dictates whether the disease is affecting one eye or both (unilateral or bilateral) [6].

Limbal stem cells transplantation

The aim of treatment for LSCD is to re-establish the physiologic and anatomic environment of the ocular surface by reconstruction of the corneal and conjunctival epithelium [36]. Some techniques to replace limbal stem cells have been reported. Currently, the main clinical procedures that are performed include CLAU, SLET, KLAL, and cultivated limbal stem cell transplantation (CLET) [51]. The source of stem cell in unilateral disorders is the healthy contralateral eye [4,52]. Subsequently, transplantation of allogeneic limbal SCs that are dissected from cadaveric donors or living tissue-matched eyes to treat bilateral LSCD [53,54]. Limbal deficiency in the donor eye is a potential serious risk to this procedure. Both of auto or allograft limbal tissues in transplantation have risks and benefits. Lid pathology, dry eye, uncontrolled systemic disorders and other different factors affect the achievement of LSC transplantation. Limbal transplantation is a definitive treatment for LSCD and may ameliorate the visual acuity of a patient with ocular surface disease [55]. The second strategy to treat LSCD is ex vivo expansion of limbal stem cells from a single small biopsy to transplant amniotic membrane (AMT), which forms the inner wall of the membranous sac surrounding the embryo during gestation. Anti-inflammatory and anti-angiogenic properties of amnion cause to use it in ocular surface reconstruction [56]. A number of clinical studies have shown transplantation of autologous, allogeneic limbal tissue or expanded cells in concomitance with AMT to promote the rapid re-epithelization required to restore corneas with LSCD [57-59]. In unilateral cases with total LSCD, transplantation of ex vivo cultured LSCs from the human eye (CLET) or a CLAU is utilized (conjunctival limbal auto-explant). Alloexplant limbal transplant performed in patients with bilateral total LSCD and it is extracted from a cadaveric or a living relative donor and then expanded in ex vivo. Recently, SLET has gained popularity in unilateral LSCD; in this technique, several pieces of one clock hour of contralateral healthy limbus are spread on a layer of AMT on the involved ocular surface [57-59].

Clinical trials of limbal stem cells transplantation:Variation of culture techniques, case selection between studies, performance of both autologous, allogeneic transplants in studies, not including the corneal surface, visual improvement in some studies and different follow-up periods limit the interpretation of clinical trial results from cultured LSC therapy [60-65]. Over the years, many different methods have been developed. The first stem cell autograft using conjunctival-limbal-corneal epithelium harvested from the healthy fellow eyes of patients with unilateral chemical burns was reported by Barraquer in 1965 [60]. In 1997, Pellegrini et al. [61] reported the first experience of the clinical use of ex vivo cultured limbal epithelial stem cells (LESCs) for treating corneal LESC deficiency. In 2003,Sangwan and colleagues published a study demonstrated reconstruction of the ocular surface in a case of severe bilateral partial LSCD with using autologous cultured conjunctival and limbal epithelium extracted from the healthy eye [62]. In addition, in 2006, they reported the clinical outcome of autologous cultivated limbal epithelial transplantation between March 2001 and May 2003.There were 88 eyes of 86 patients with limbal stem cell deficiency (LSCD). Sixty-four percent of patients were due to alkali burns and 69% eyes had total LSCD. Finally results shown 73.1% (57 eyes) were successful outcome with a stable ocular surface without conjunctivalization, 26.9% (21 eyes) had a considered failures and 10 patients were lost to follow-up [63]. The purpose of the study performed by Sangwan et al in 2011 was the efficacy of xeno-free autologous cell-based treatment with unilateral total limbal stem cell deficiency due to ocular surface burns treated between 2001 and 2010. A small limbal biopsy was taken from the healthy eye and the limbal epithelial cells were expanded ex vivo on human amniotic membrane using a xeno-free explant culture system. The resulting cultured epithelial monolayer and amniotic membrane substrate were transplanted on to the patient’s affected eye. A completely epithelized, avascular and clinically stable corneal surface was seen in142 of 200 (71%) eyes in this retrospective study. An improvement in visual acuity, without further surgical intervention, was seen in 60.5% of eyes. All donor eyes remained healthy [64]. In 2005, Daya performed a study on 10 eyes of 10 patients with profound LSCD due to ectodermal dysplasia (3 eyes), Stevens-Johnson syndrome (3 eyes), chemical injury (2 eyes), thermal injury (1 eye), and rosacea blepharoconjunctivitis (1 eye) to investigate the outcome of ex vivo expanded stem cell allograft for LSCD. Seventy percent of eyes (7 of 10) had improved parameters, including vascularization, conjunctivalization, inflammation, epithelial defect, photophobia, and pain and 40% of eyes (4 cases of 7) had improved visual acuity [66]. A study by Nakamuraet al (2006) investigated 9 eyes from 9 patients with total limbal stem cell deficiency (2 eyes with Stevens-Johnson syndrome, 1 with chemical injury, 1 with ocular cicatricial pemphigoid, 1 with Salzmann corneal dystrophy, 1 with aniridia, 1 with graft-versus-host disease, and 2 with idiopathic ocular surface disease). The authors compared autologous serum (AS)-derived corneal epithelial equivalents with those derived from fetal bovine serum (FBS)-supplemented medium, so cultivated corneal epithelial transplantation can be used for the treatment of severe ocular surface disease. Allologous (7 cases) and autogenic (2 cases) AS-derived cultivated corneal epithelial equivalents were transplanted onto the ocular surface. During the follow-up period, the corneal surface of all patients remained stable and transparent, without significant complications and visual acuity improvement was seen in all eyes [67]. In 2006, Javadi et al. published a more detailed report on the early results of transplantation of autologous limbal stem cells cultivated on amniotic membrane (AM) in four eyes of 4 patients with total unilateral LSCD. After 5-13-month follow-up, visual acuity, corneal opacification, and vascularization improved in all cases [68]. In 2010, they performed keratolimbal allograft (KLAL) for treatment of 21 eyes of 20 patients with total LSCD and adequate tear production were included. Mean visual acuity improved. Graft survival rate was 61.9% at 1 year and 31% at 20 months [69]. Alex J. Shortt used a novel culture system without 3T3 feeder cells to determine the outcome of ex vivo cultured LESC transplantation. Allogeneic (7 eyes) and autologous (3 eyes) corneal LESCs were cultured on human amniotic membrane. Tissue was transplanted to the recipient eye after superficial keratectomy. The success rate was 60% with a successful outcome experienced [70]. In 2014, Vazirani with the cooperation of Sangwan performed a study on seventy eyes of 70 patients with unilateral, partial LSCD and reported the outcomes of autologous cultivated limbal epithelial transplantation using the healthy part of the affected eye or the fellow eye as a source of limbal stem cells in patients. In 36 eyes, the limbal biopsy was harvested from the healthy fellow eye (contralateral group) and in the remaining eyes from the healthy part of the limbus of the same eye (ipsilateral group). Clinical success was achieved in 70.59% of eyes in the ipsilateral group and 75% of eyes in the contralateral group. Outcomes are similar irrespective of whether the limbal biopsy is taken from the healthy part of the ipsilateral eye or the contralateral eye [65].

Stem cell therapy for retinal degenerative diseases

Introduction:Every year many people suffer from visual disturbance or even blindness caused by retinal detachment. The retina is a part of the central nervous system (CNS) consisting of neuronal cells (photoreceptors, horizontal cells, amacrine cells, bipolar cells and retinal ganglion cells) and glial cells (Müller glia is a specialized type of glial cell only present in the retina) [71]. Epidemiologists have found that damage to the retina can occur the entire age spectrum. For instance, the pediatric and young adult populations are affected by retinitis pigmentosa (RP) and middle-aged adults are affected by diabetic retinopathy (DR), and the elderly are affected by age related macular degeneration (AMD) [72,73]. Retinal neurodegenerative disorders divided into diseases affecting the inner retina, example glaucoma and can affect both bipolar cells and retinal ganglion cells (RGCs) [74]. Those affecting the outer retina often leads to the death of the photoreceptors. Retinal progenitor cells were identified as possible cell candidates for an accepted potential treatment strategy for retinal injury. These cells represent many of the properties associated with stem cells: (1) proliferation and expression of Nestin, a neuroectodermal stem cell marker, (2) multipotential property, and (3) self-renewal capacity [75]. Photoreceptors, intermediate neurons and Muller glia are differentiated in vitro to form sphere colonies of cell. Progenitor cells find themselves in an inhibitory environment and this is the result of the failure of retinal progenitor cells to renew retinal cells in the postnatal period [76]. Muller glia cells play a key role in the generation of multipotent precursor cells from embryonic retinal cells and these have the potential to become neurogenic retinal progenitor cells [77]. In retinal disease such as retinitis pigmentosa and age-related macular degeneration can use retinal progenitor cells for transplantation in the adult retina. Main sources of progenitor cells are: embryo, the bone marrow, neuronal genesis region, and eye (ciliary body epithelium, the iris, the ciliary marginal zone, and retina) [78].

Stem cells in retina:The ability of donor cells migrate into the desired location, to be alive after transplantation, and to differentiate into retinal cells are the three causes successful of stem cell therapy. Recent researches have shown embryonic SC (ESC), adult SC and induced pluripotent SC (iPSC) are three main types of stem cells being considered as the potential source for retinal repair and regeneration. These eye-derived PCs have the potential to differentiate into retina-specific cells in an allowable environment. IPSCs are somatic cells, which can be genetically reprogrammed to become ESC-like with the risk of tumor genesis. A sub-population of Muller glia with SC characteristics has been founded in the adult human retina. The aim of transplantation of functional retinal cells or stem cells is to restore vision by repopulating the degenerated retina via rescuing retinal neurons from further degeneration [79-81].

Target cell types for retinal degeneration treatment

Retinal pigmented epithelial cells (RPE):Human embryonic stem cells (HESCs) and human induced pluripotent stem cells (hiPSCs) can be differentiated into all retinal cell types [82] RPE plays a key role in maintenance of neural retinal function that can suggest retinal degeneration can be treated with sub-retinal injections of RPE cells. The improvement in stem cell differentiation techniques can make pluripotent stem cells differentiation into RPE cells. Extended monolayers of RPE can be isolated and transferred to a variety of substrates [83-85].

Photoreceptor:The retina includes highly specialized photoreceptors that capture the photons and transduce them into electrical signals. The ability to produce true multipotent neuroretinal progenitor cells (NRPCs) and being renewable are distinct advantages of human pluripotent stem cells (HPSCs) as sources of donor neuroretinal cell types [86]

Discussion

The focus of this review was principal stem cells in the eye especially limbal stem cells. One of the most important ocular stem cells is limbal stem cell that retains in an undifferentiated state and exists in an optimal microenvironment or “niche”. The importance of limbal stem cells in maintaining the corneal epithelium throughout life has long been recognized. Moreover, stem cells in the retina as possible edge of the science, have been suggested for treatment of the retinal degeneration, which generally results in constant visual disturbance or even blindness. However, the limbal stem cells have been studied deeply during previous days. When the patient is labeled as LSCD according to the clinical features, one of above-mentioned procedures could be done. In bilateral cases a KLAL or CLET procedure may be needed. In unilateral involvement, a CLAU or SLET could be savior. It seems that we need more studies in the field of ophthalmic stem cells

1 Pellegrini G, De Luca M, Arsenijevic Y (2007) Towards therapeutic application of ocular stem cells. Semin Cell Dev Biol 18: 805-18.
2 Dua HS, Azuara-Blanco A (2000) Limbal stem cells of the corneal epithelium. Surv Ophthalmol 44: 415-25.
3 Shanbhag SS, Saeed HN, Paschalis EI, Chodosh J (2017) Keratolimbal allograft for limbal stem cell deficiency after severe corneal chemical injury: a systematic review. Bjophthalmol 311249.
4 Dua HS, Azuara-Blanco A (2000) Autologous limbal transplantation in patients with unilateral corneal stem cell deficiency. Br J Ophthalmol 84: 273-8.
5 Sepsakos L, Cheung AY, Holland EJ (2017) Outcomes of Keratoplasty after Ocular Surface Stem Cell Transplantation. Cornea 36: 1025-30.
6 Ahmad S, Kolli S, Lako M, Figueiredo F, Daniels JT (2010) Stem cell therapies for ocular surface disease. Drug Discov Today 15: 306-13.
7 Ramos T, Scott D, Ahmad S (2015) An update on ocular surface epithelial stem cells: cornea and conjunctiva. Stem Cells Int 2015: 601731.
8 Le Q, Xu J, Deng SX (2018) The diagnosis of limbal stem cell deficiency. Ocul Surf 16: 58-69.
9 Fuchs E, Tumbar T, Guasch G (2004) Socializing with the neighbors: stem cells and their niche. Cell 116: 769-78.
10 Chen SY, Han B, Zhu YT, Mahabole M, Huang J, et al. (2015) HC-HA/PTX3 Purified From Amniotic Membrane Promotes BMP Signaling in Limbal Niche Cells to Maintain Quiescence of Limbal Epithelial Progenitor/Stem Cells. Stem Cells 33: 3341-55.
11 Romano AC, Espana EM, Yoo SH, Budak MT, Wolosin JM, et al. (2003) Different cell sizes in human limbal and central corneal basal epithelia measured by confocal microscopy and flow cytometry. Invest Ophthalmol Vis Sci 44: 5125-9.
12 Basu S, Bahuguna C, Singh V (2018) Simple limbal epithelial transplantation: Impactful innovation. Indian J Ophthalmol 66: 53-4.
13 Davies SB, Chui J, Madigan MC, Provis JM, Wakefield D, et al. (2009) Stem cell activity in the developing human cornea. Stem Cells 27: 2781-92.
14 Shortt AJ, Secker GA, Munro PM, Khaw PT, Tuft SJ (2007) Characterization of the limbal epithelial stem cell niche: novel imaging techniques permit in vivo observation and targeted biopsy of limbal epithelial stem cells. Stem Cells 25: 1402–9.
15 Tseng SC, He H, Zhang S, Chen SY (2016) Niche Regulation of Limbal Epithelial Stem Cells: Relationship between Inflammation and Regeneration. Ocul Surf 14: 100-12.
16 Watt FM, Hogan BL (2000) Out of Eden: stem cells and their niches. Science 287: 1427-30.
17 Fuchs E, Tumbar T, Guasch G (2004) Socializing with the neighbors: stem cells and their niche. Cell 116: 769-78.
18 Cotsarelis G (2006) Epithelial stem cells: a folliculocentric view. J Invest Dermatol 126: 1459-68.
19 Blanpain C, Horsley V, Fuchs E (2007) Epithelial Stem Cells: Turning over New Leaves. Cell 128: 445-58.
20 Ahmad S, Stewart R, Yung S, Kolli S, Armstrong L, et al. (2007) Differentiation of Human Embryonic Stem Cells into Corneal Epithelial‐Like Cells by In Vitro Replication of the Corneal Epithelial Stem Cell Niche. Stem Cells 25: 1145-55.
21 Kim KH, Mian SI (2017) Diagnosis of corneal limbal stem cell deficiency. Curr Opin Ophthalmol 28: 355-362.
22 Vazirani J, Nair D, Shanbhag S, Wurity S, Ranjan A (2018) Limbal Stem Cell Deficiency - Demography And Underlying Causes. Am J Ophthalmol 9394: 30026- 6.
23 Schlötzer-Schrehardt U, Kruse FE (2005) Identification and characterization of limbal stem cells. Exp Eye Res 81: 247-64.
24 O’Sullivan F, Clynes M (2007) Limbal stem cells, a review of their identification and culture for clinical use. Cytotechnology 53: 101-6.
25 Kubota M, Shimmura S, Miyashita H, Kawashima M, Kawakita T, et al. (2010) The anti-oxidative role of ABCG2 in corneal epithelial cells. Invest Ophthalmol Vis Sci 51: 5617-22.
26 Zhou S, Schuetz JD, Bunting KD, Colapietro AM, Sampath J, et al. (2001) The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med 7: 1028-34.
27 Umemoto T, Yamato M, Nishida K, Yang J, Tano Y, et al. (2006) Limbal Epithelial Side‐Population Cells Have Stem Cell–Like Properties, Including Quiescent State. Stem Cells 24: 86-94.
28 Lohrum MA, Vousden KH (2000) Regulation and function of the p53-related proteins: same family, different rules. Trends Cell Biol 10: 197-202.
29 Celli J, Duijf P, Hamel BC, Bamshad M, Kramer B, et al. (1999) Heterozygous germline mutations in the p53 homolog p63 are the cause of EEC syndrome. Cell 99: 143-53.
30 Barbaro V, Testa A, Di Iorio E, Mavilio F, Pellegrini G, et al. (2007) C/EBPδ regulates cell cycle and self-renewal of human limbal stem cells. J Cell Biol 177: 1037-49.
31 Ahmad S, Figueiredo F, Lako M (2006) Corneal epithelial stem cells: characterization, culture and transplantation. Regen Med 1: 29-44.
32 Nakamura T, Inatomi T, Sotozono C, Koizumi N, Kinoshita S (2016) Ocular surface reconstruction using stem cell and tissue engineering. Prog Retin Eye Res 51: 187-207.
33 Takács L, Tóth E, Berta A, Vereb G (2009) Stem cells of the adult cornea: from cytometric markers to therapeutic applications. Cytometry A 75: 54-66.
34 Thomas PB, Liu YH, Zhuang FF, Selvam S, Song SW, et al. (2007) Identification of Notch-1 expression in the limbal basal epithelium. Mol Vis 13: 337-44.
35 Whitcher JP, Srinivasan M, Upadhyay MP (2001) Corneal blindness: a global perspective. WHO 79: 214-21.
36 Bakhtiari P, Djalilian A (2010) Update on limbal stem cell transplantation. Middle East Afr J Ophthalmol 17: 9-14.
37 Li W, Chen YT, Hayashida Y, Blanco G, Kheirkah A, et al. (2008) Down‐regulation of Pax6 is associated with abnormal differentiation of corneal epithelial cells in severe ocular surface diseases. J Pathol 214: 114-22.
38 Dhamodaran K, Subramani M, Jeyabalan N, Ponnalagu M, Chevour P, et al. (2015) Characterization of ex vivo cultured limbal, conjunctival, and oral mucosal cells: A comparative study with implications in transplantation medicine. Mol Vis 21: 828-45.
39 Puangsricharern V, Tseng SC (1995) Cytologlogic evidence of corneal diseases with limbal stem cell deficiency. Ophthalmol (10): 1476-85.
40 Kruse FE (1994) Stem cells and corneal epithelial regeneration. Eye 8: 170-83.
41 Pfister RR (1994) Corneal stem cell disease: concepts, categorization, and treatment by auto-and homotransplantation of limbal stem cells. Eye & Contact Lens 20: 64-72.
42 Fujishima H, Shimazaki J, Tsubota K (1996) Temporary corneal stem cell dysfunction after radiation therapy. Br J Ophthalmol 80: 911-4.
43 Dua HS, Joseph A, Shanmuganathan VA, Jones RE (2003) Stem cell differentiation and the effects of deficiency. Eye 17: 877-85.
44 Lavker RM, Tseng SC, Sun TT (2004) Corneal epithelial stem cells at the limbus: looking at some old problems from a new angle. Exp Eye Res 78: 433-46.
45 Tseng SC, Sun TT (1999) Stem cells: ocular surface maintenance. Corneal surgery theory, technique, tissue. St. Louis 9-18.
46 Puangsricharern V, Tseng SC (1995) Cytologlogic evidence of corneal diseases with limbal stem cell deficiency. Ophthalmol 102: 1476-85.
47 Tseng SC (1996) Regulation and clinical implications of corneal epithelial stem cells. Mol Biol Rep 23: 47-58.
48 Huang AJ, Tseng SC (1991) Corneal epithelial wound healing in the absence of limbal epithelium. Invest Ophthalmol Vis Sci 32: 96-105.
49 Shortt AJ, Secker GA, Notara MD, Limb GA, Khaw PT, et al. (2007) Transplantation of ex vivo cultured limbal epithelial stem cells: a review of techniques and clinical results. Surv Ophthalmol. 52: 483-502.
50 Coster DJ, Aggarwal RK, Williams KA (1995) Surgical management of ocular surface disorders using conjunctival and stem cell allografts. Br J Ophthalmol 79: 977-82.
51 Holland EJ, Schwartz GS (2004) The Paton lecture: ocular surface transplantation: 10 years’ experience. Cornea 23: 425-31.
52 Tsai RJ, Tseng SC (1994) Human allograft limbal transplantation for corneal surface reconstruction. Cornea 13: 389-400.
53 Dua HS, Azuara-Blanco A (2000) Limbal stem cells of the corneal epithelium. Surv Ophthalmol 44: 415-25.
54 Holland EJ (1996) Epithelial transplantation for the management of severe ocular surface disease. Trans Am Ophthalmol Soc 94: 677.
55 Nishiwaki-Dantas MC, Dantas PE, Reggi JR (2001) Ipsilateral limbal translocation for treatment of partial limbal deficiency secondary to ocular alkali burn. Br J Ophthalmol 85: 1031-3.
56 Tsai RJ, Li LM, Chen JK (2000) Reconstruction of damaged corneas by transplantation of autologous limbal epithelial cells. Am J Opthamol 4: 543.
57 Basu S, Sureka SP, Shanbhag SS, Kethiri AR, Singh V, et al. (2016) Simple Limbal Epithelial Transplantation: Long-Term Clinical Outcomes in 125 Cases of Unilateral Chronic Ocular Surface Burns. Ophthalmology 123: 1000-10.
58 Tseng SC, Prabhasawat P, Barton K, Gray T, Meller D (1998) Amniotic membrane transplantation with or without limbal allografts for corneal surface recon- struction in patients with limbal stem cell deficiency. Ophthalmology 116: 431-41.
59 Mittal V, Jain R, Mittal R, Vashist U, Narang P (2016) Successful management of severe unilateral chemical burns in children using simple limbal epithelial transplantation (SLET). Br J Ophthalmol 100: 1102-8.
60 Barraquer RI, de Toledo JP, Barraquer J (1997) Storage, surgery, outcome, complications, and new developments in corneal and conjunctival grafts. Curr Opin Ophthalmol 8: 31-40.
61 Pellegrini G, Traverso CE, Franzi AT, Zingirian M, Cancedda R, De Luca M (1997) Long-term restoration of damaged corneal surfaces with autologous cul- tivated corneal epithelium. Lancet 349: 990-3.
62 Sangwan VS, Vemuganti GK, Iftekhar G, Bansal AK, Rao GN (2003) Use of autologous cultured limbal and conjunctival epithelium in a patient with severe bilateral ocular surface disease induced by acid injury: a case report of unique application. Cornea 22: 478-81.
63 Sangwan VS, Matalia HP, Vemuganti GK, Fatima A, Ifthekar G, et al. Clinical outcome of autologous cultivated limbal epithelium transplantation. Indian J Ophthalmol 54: 29-34.
64 Sangwan VS, Basu S, Vemuganti GK, Sejpal K, Subramaniam SV, et al. (2011) Clinical outcomes of xeno-free autologous cultivated limbal epithelial transplanta- tion: a 10-year study. Br J Ophthalmol 95: 1525-9.
65 Vazirani J, Basu S, Kenia H, Ali MH, Kacham S, et al. (2014) Unilateral partial limbal stem cell deficiency: contralateral versus ipsilateral autologous cultivated limbal epithelial transplantation. Am J Ophthalmol 157: 584-90.
66 Daya SM, Watson A, Sharpe JR, Giledi O, Rowe A, et al. (2005) Outcomes and DNA analysis of ex vivo expanded stem cell allograft for ocular surface recon- struction. Ophthalmology 112: 470-7.
67 Nakamura T, Inatomi T, Sotozono C, Ang LP, Koizumi N, et al. (2006) Transplantation of autologous serum-derived cultivated corneal epithelial equivalents for the treatment of severe ocular surface disease. Ophthalmology 113: 1765-72.
68 Javadi MA, Einollahi B, Rafiei AB, Baharvand H, Ebrahimi M, et al (2006) Early results of autologous cultivated limbal stem cell transplantation in total limbal stem cell deficiency. Iranian J Ophthalmic Res 1: 71-80.
69 Baradaran-Rafii AR, Abdolahi A, Jabarpoor Bonyadi MH, Mirdehghan SA, Javadi MA, et al. (2010) Keratolimbal allograft in total limbal stem cell deficiency. Bina J Ophthalmol 15: 167-76.
70 Shortt AJ, Secker GA, Rajan MS, Meligonis G, Dart JK, et al. (2008) Ex vivo expansion and transplantation of limbal epithelial stem cells. Ophthalmology 115: 1989-97.
71 Bull ND, Martin KR (2011) Concise review: Toward stem cell‐based therapies for retinal neurodegenerative diseases. Stem Cells 29: 1170-5.
72 Sohocki MM, Daiger SP, Bowne SJ, Rodriquez JA, Northrup H, et al. (2001) Prevalence of mutations causing retinitis pigmentosa and other inherited retinopa- thies. Hum Mutat 17: 42-51.
73 Kobrin Klein BE (2007) Overview of epidemiologic studies of diabetic retinopathy. Ophthalmic Epidemiol 14: 179-83.
74 Johnson TV, Bull ND, Hunt DP, Marina N, Tomarev SI, et al. (2010) Neuroprotective effects of intravitreal mesenchymal stem cell transplantation in experimen- tal glaucoma. Investi Ophthalmo Vis Sci 51: 2051-9.
75 Fischer AJ, Reh TA (2003) Potential of Müller glia to become neurogenic retinal progenitor cells. Glia 43: 70-6.
76 Mayer EJ, Hughes EH, Carter DA, Dick AD (2003) Nestin positive cells in adult human retina and in epiretinal membranes. Brit J Ophthalmol 87: 1154-8.
77 Layer PG, Rothermel A, Willbold E (2001) From stem cells towards neural layers: a lesson from re-aggregated embryonic retinal cells. Neuroreport 12: A39-46.
78 Bi YY, Feng DF, Pan DC (2009) Stem/progenitor cells: a potential source of retina-specific cells for retinal repair. Neuroscience research 65: 215-21.
79 Tropepe V, Coles BL, Chiasson BJ, Horsford DJ, Elia AJ, et al. (2000) Retinal stem cells in the adult mammalian eye. Science 287: 2032-6.
80 Coles BL, Angénieux B, Inoue T (2004) Facile isolation and the characterization of human retinal stem cells. Proc Natl Acad Sci U S A 101: 15772-7.
81 Chacko DM, Rogers JA, Turner JE, Ahmad I (2000) Survival and differentiation of cultured retinal progenitors transplanted in the subretinal space of the rat. Biochem Biophy Res Comm 268: 842-6.
82 Rowland TJ, Buchholz DE, Clegg DO (2012) Pluripotent human stem cells for the treatment of retinal disease. J Cell Physiol 227: 457-66.
83 Meyer JS, Howden SE, Wallace KA, Verhoeven AD, Wright LS, et al. (2011) Optic vesicle‐like structures derived from human pluripotent stem cells facilitate a customized approach to retinal disease treatment. Stem cells 29: 1206-18.
84 Buchholz DE, Hikita ST, Rowland TJ, Friedrich AM, Hinman CR, et al. (2009) Derivation of functional retinal pigmented epithelium from induced pluripotent stem cells. Stem cells 27: 2427-34.
85 Singh R, Shen W, Kuai D, Martin JM, Guo X, et al. (2012) iPS cell modeling of Best disease: insights into the pathophysiology of an inherited macular degenera- tion. Human molecular genetics 22: 593-607.
86 Borooah S, Phillips MJ, Bilican B (2013) Using human induced pluripotent stem cells to treat retinal disease. Progress in Retinal Eye Res 37: 163-81.
87 van Meurs JC, Van Den Biesen PR (2003) Autologous retinal pigment epithelium and choroid translocation in patients with exudative age-related macular degeneration: short-term follow-up. Am J Ophthalmol 136: 688-95.
88 Otani A, Dorrell MI, Kinder K, Moreno SK, Nusinowitz S, et al. (2004) Rescue of retinal degeneration by intravitreally injected adult bone marrow–derived lineage-negative hematopoietic stem cells. J Clin Invest 114: 765-774.
89 Wang S, Lu B, Wood P, Lund RD (2005) Grafting of ARPE-19 and Schwann cells to the subretinal space in RCS rats. Invest Ophthalmo Vis Sci 46: 2552-60.
90 Lund RD, Wang S, Klimanskaya I, Holmes T, Ramos-Kelsey R, (2006) Human embryonic stem cell–derived cells rescue visual function in dystrophic RCS rats. Cloning and stem cells 8: 189-99.
91 MacLaren RE, Uppal GS, Balaggan KS, Tufail A, Munro PM, et al. (2007) Autologous transplantation of the retinal pigment epithelium and choroid in the treat- ment of neovascular age-related macular degeneration. Ophthalmol 114: 561-70.
92 Castanheira P, Torquetti L, Nehemy MB, Goes AM (2008) Retinal incorporation and differentiation of mesenchymal stem cells intravitreally injected in the injured retina of rats. Arq Bras Oftalmol 71: 644-50.
93 Lu B, Malcuit C, Wang S, Girman S, Francis P, et al. (2009) Long‐term safety and function of RPE from human embryonic stem cells in preclinical models of macular degeneration. Stem cells 27: 2126-35.
94 Levkovitch-Verbin H, Sadan O, Vander S, et al. (2010) Intravitreal injections of neurotrophic factors secreting mesenchymal stem cells are neuroprotective in rat eyes following optic nerve transection. Invest Ophthalmo Vis Sci 51: 6394-400.
95 West EL, Pearson RA, Barker SE, Luhmann UF, Maclaren RE, et al. (2010) Long‐term survival of photoreceptors transplanted into the adult murine neural retina requires immune modulation. Stem cells 28: 1997-2007.
96 Takeuchi K, Kachi S, Iwata E, Ishikawa K, Terasaki H (2012) Visual function 5 years or more after macular translocation surgery for myopic choroidal neovas- cularisation and age-related macular degeneration. Eye 26: 51-60.
97 Falkner-Radler CI, Krebs I, Glittenberg C, Povazay B, Drexler W, et al. (2010) Human retinal pigment epithelium (RPE) transplantation: outcome after autolo- gous RPE-choroid sheet and RPE cell-suspension in a randomised clinical study. Br J Ophthalmol 95: 370-5.
98 Schwartz SD, Hubschman JP, Heilwell G, Franco-Cardenas V, Pan CK, et al. (2012) Embryonic stem cell trials for macular degeneration: a preliminary report. The Lancet 379: 713-20.
99 Hu Y, Liu L, Lu B, Zhu D, Ribeiro R, et al. (2012) A novel approach for subretinal implantation of ultrathin substrates containing stem cell-derived retinal pig- ment epithelium monolayer. Ophthalmic Res 48: 186-91.
100 Homma K, Okamoto S, Mandai M, Gotoh N, Rajasimha HK, et al. (2013) Developing Rods Transplanted into the Degenerating Retina of Crx‐Knockout Mice Exhibit Neural Activity Similar to Native Photoreceptors. Stem Cells 31: 1149-59.
101 Buchholz DE, Pennington BO, Croze RH, Hinman CR, Coffey PJ, et al. (2013) Rapid and efficient directed differentiation of human pluripotent stem cells into retinal pigmented epithelium. Stem Cells Transl Med 2: 384-93.
102 Kamao H, Mandai M, Okamoto S, Sakai N, Suga A, et al. (2014) Characterization of human induced pluripotent stem cell-derived retinal pigment epithelium cell sheets aiming for clinical application. Stem Cell Rep 2: 205-18.
103Tzameret A, Sher I, Belkin M, Treves AJ, Meir A, et al. (2014) Transplantation of human bone marrow mesenchymal stem cells as a thin subretinal layer ame- liorates retinal degeneration in a rat model of retinal dystrophy. Exp Eye Res 118: 135-44.
104 Pennington BO, Clegg DO, Melkoumian ZK, Hikita ST (2015) Defined culture of human embryonic stem cells and xeno-free derivation of retinal pigmented epithelial cells on a novel, synthetic substrate. Stem Cells Transl Med 4: 165-77.
105 Chen Z, Zhang YA (2015) Cell therapy for macular degeneration--first phase I/II pluripotent stem cell-based clinical trial shows promise. Sci China Life Sci 58: 119.

Journal of Stem Cells and Clinical Practice

Tables at a glance
table-icon
Table 1
table-icon
Table 2

Markers

 

 

ABCG2 [25-27]

ATP binding cassette transporter protein, in the limbal basal epithelium, 0.3-0.5% of cells in the limbal epithelium exhibit the side-population phenotype, to protect LSCs against oxidative stress induced by toxins

 

p63 [28-30]

a transcription factor, potential keratinocyte stem cell marker, DNp63α* is thought to be a highly expressed in the limbus during resting state, important for epithelial development

C/EBPδ [30,32]

CCAAT enhancer binding protein delta, a transcription factor, induces G0/G1 cell cycle arrest in mammary gland epithelial cells,

 

Bmi1 [33]

a repressor that to be expressed in the limbal epithelial side- population, a Polycomb group repressor involved in the self- renewal of various types of adult stem cell

 

Notch 1 [34]

a transmembrane receptor, maintaining cells in an undifferentiated state, localized to a small number of cells in the limbal epithelial basal layer, co-expressed with ABCG2

DNp63a is one of 3 isoforms of p63 without an added transactivation domain (There are 6 isoforms of p63) has been shown to be more specific for LESCs than the other isoforms. [31] LSC: limbal stem cell. ATP: adenosine triphosphate
Table 1: Important stem cell markers

Study

Retina disease

Outcomes

2003

 

 

Van Meurs JC et al. [87]

AMD (clinical)

Postoperative vision with a 2-line increase in three patients

2004

 

 

Otani A et al. [88]

Retinal degeneration

Treatment

2005

 

 

Wang S et al. [89]

Photoreceptor loss

Survive and rescue photoreceptors using grafts

2006

 

 

Lund RD et al. [90]

Retinal dystrophy

Improvement in visual performance was 100%

2007

 

 

MacLaren RE et al. [91]

neovascular AMD (clinical)

Autologous RPE transplantation restores vision

2008

 

 

Castanheira P et al. [92]

Retinal degeneration

Rod photoreceptor, bipolar and amacrine cell markers were expressed by grafted cells

2009

 

 

Buchholz DE et al. [84]

Retinitis pigmentosa and AMD

iPSCs differentiate into functional RPEs

Lu B et al. [93]

Macular degeneration

The cells sustained visual function and photoreceptor integrity

2010

 

 

Levkovitch-Verbin H et al. [94]

Glaucoma

BM-MSCs deliver neurotrophic factors and neuroprotection

West EL et al. [95]

Neural retina repair

Photoreceptors were present up to 12 months post-transplantation

2011

 

 

Takeuchi K etal. [96]

AMD

Maintenance of visual acuity

Falkner-Radler et al. [97]

AMD

Autologous RPE sheet, Maintenance of visual acuity

2012

 

 

 

Schwartz etal. [98]

 

AMD

Protection of retina in macular degeneration by replacement of the structural and trophic support provided by retinal pigment epithelium

Hu Y et al. [99]

RCS rat

Successful transplantation of hESC- RPE graft patch

2013

 

 

K Homma et al. [100]

AMD and retiniti pigmentosa

Neural activity similar to native photoreceptors

Buchholz D et al. [101]

retinitis pigmentosa

HiPSCs less efficient in comparison with hESC

2014

 

 

 

Hiroyuki Kamao et al. [102]

 

AMD

RPE cell sheets generated without any artificial cell sheets and performed subretinal injection into RCS

Adi Tzameret etal. [103]

retinal dystrophy

rescue of retinal function and significantly delayed photoreceptor degeneration

2015

 

 

 

Britney O et al. [104]

 

AMD

hESCs and their differentiation maintaining into RPE using Xeno-Free derivation (a novel, synthetic substrate)

Chen ZG et al. [105]

AMD and Stargardt’s macular dystrophy

increase in visual acuity

Table 2: Recent studies on stem cell therapy in retinal diseases. AMD: age related macular degeneration, RPE: retinal pigment epithelium, hESC: human embryonic stem cell, RCS: Royal College of Surgeons

Partnered Content Networks

  • Cancer Science
  • Vaccine Studies
  • Gynecology
  • Food Nutrition
  • Nursing Science
  • Public Health
  • The Pharma
  • Infectious Disease
  • Neuro Care
  • Catalysis
  • Neonatal Biology
  • Neonatal Disorders
  • Mutation
  • Nanotechnology
  • Toxicology
  • Dark Biotechnology
  • Pollution Toxicology
  • Cell Biology
  • Bioanalytical Research
  • Renal Disorders
  • The Astrophysics
  • Sleep Physiology
  • Epidemiology
  • Histology