Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Stem Cell Rev (2008) 4:159–168

DOI 10.1007/s12015-008-9029-x

Corneal Epithelial Stem Cells: Deficiency and Regulation


Genevieve A. Secker & Julie T. Daniels

Published online: 12 July 2008


# Humana Press 2008

Abstract The corneal epithelium is continuously renewed Limbal Epithelial Stem Cells
by a population of stem cells that reside in the corneoscleral
junction, otherwise known as the limbus. These limbal All self-renewing tissue must contain a stem cell pool,
epithelial stem cells (LESC) are imperative for corneal which provides an unlimited supply of proliferating cells.
maintenance with deficiencies leading to in-growth of This is true for the corneal epithelium with a large body of
conjunctival cells, neovascularisation of the corneal stroma research indicating these cells reside in the limbal basal
and eventual corneal opacity and visual loss. One such region and are aptly named limbal epithelial stem cells
disease that has traditionally been thought to be due to (LESC; Fig. 1). LESC share a number of features with
LESC deficiency is aniridia, a pan-ocular congenital eye other adult somatic stem cells. These include having small
disease due to mutations in the PAX6 gene. Corneal changes cell size [1], the lack expression of differentiation markers,
or aniridia related keratopathy (ARK) seen in aniridia are such as cytokeratin 3/12 [2, 3] and high nuclear to
typical of LESC deficiency. However, the pathophysiology cytoplasmic ratio [7].
behind ARK is still ill defined, with current theories LESC are considered to be primitive cells as they are
suggesting it may be caused by a deficiency in the stem slow cycling and therefore label retaining under normal
cell niche and adjacent corneal stroma, with altered wound conditions but have the ability to be highly proliferative in
healing responses also playing a role (Ramaesh et al, response to injury [5–7]. Stem cells have the ability to
International Journal of Biochemistry & Cell Biology divide asymmetrically to repopulate the stem cell pool.
37:547–557, 2005) or abnormal epidermal differentiation Barbaro et al., recently found expression of C/EBPδ in a
of LESC (Li et al., The Journal of Pathology 214:9, 2008). subset of LESC both in vivo and in vitro, and have
PAX6 is considered the master control gene for the eye and suggested it is involved in the regulation of self renewal
is required for normal eye development with expression and cell cycle length of LESC. Other pathways have been
continuing in the adult cornea, thus inferring a role for linked to stem cell renewal, such as Notch-1. Corneal
corneal repair and regeneration (Sivak et al., Developments specific inducible ablation of Notch1 demonstrated differ-
in Biologicals 222:41–54, 2000). Studies of models of Pax6 entiation of LESC into hyperplastic, keratinised skin like
deficiency, such as the small eyed (sey) mouse, should help epithelium [8]. Furthermore LESC express progenitor
to reveal the intrinsic and extrinsic mechanisms involved in markers, including, p63 [9], ABCG2 [10] and more
normal LESC function. recently N-cadherin

Keywords Limbal epithelial stem cells . Limbus . Cornea .


Aniridia . PAX6
Evidence for the Location of LESC to the Limbus

G. A. Secker (*) : J. T. Daniels The first experimental evidence for the location of LESC to
Cells for Sight Transplantation and Research Programme, the limbus was the movement of pigment from the limbal
Ocular Repair and Regeneration Biology Unit, region towards an epithelial defect in rabbit corneas
Division of Pathology, UCL Institute of Ophthalmology,
following wounding [11]. Some years later Davanger and
11-43 Bath Street,
London EC1V 9EL, UK Evenson [12], observed a similar migration of pigment
e-mail: g.secker@ucl.ac.uk from limbus to central cornea and proposed that the
160 Stem Cell Rev (2008) 4:159–168

Fig. 1 A cross-sectional
diagram of the human corneal
limbus. Limbal epithelial stem
cells reside in the basal layer of
the epithelium (Ep) which
undulates at the limbus.
Daughter transient amplifying
cells divide and migrate towards
the central cornea (arrowed) to
replenish the epithelium, which
rests on Bowman’s layer (BL).
The stroma (St) of the limbal
epithelial stem cell niche is
populated with fibroblasts and
melanocytes and also has a
blood supply

Palisades of Vogt (PV) situated in the corneal limbus In vivo and in vitro studies have found that limbal basal
provided the source of LESC [13]. This movement from cells have a higher proliferative potential when compared to
limbal to central cornea has been described as centripetal peripheral and central cornea. Large epithelial wounds in
migration. This was demonstrated by gradual replacement rabbits have been shown to heal faster than smaller central
of donor epithelium with host cells following lamellar defects, implying that the proliferative capacity of the
keratoplasty, by looking at the dilution of sex chromatin peripheral cornea is greater than that of the central [21]. In
using a female donor graft in a male recipient in rabbits the human, limbal explant cultures have a greater prolifer-
[14]. Furthermore, the complete removal of the limbus ative potential when compared to central explants [22, 23].
results in impaired corneal function, neovascularisation and Based on human epidermal studies [4], supporting clono-
conjunctival ingrowth [13]. genicity studies found cells isolated from the limbus
As stem cells are slow cycling they divide occasionally produced the larger holoclones (stem cell derived) com-
and therefore can be identified as label retaining cells pared to the less clonogenic meroclones and paraclones
(LRCs) [15]. Exposing cells to DNA precursors such as found elsewhere in the cornea [24]. Furthermore, LESC
tritiated thymidine and bromodeoxyuridine followed by a proliferation is resistant to inhibition by tumour-promoting
chase period of 4–8weeks, the slow cycling stem cells phorbol esters [25, 26].
retain this label whereas the more differentiated transient Supplementary to experimental studies the clinical
amplifying cells (TAC) undergo dilution of the label evidence also points toward the limbus as the location of
through multiple divisions. Through the use of tritiated corneal stem cells. In normal corneal maintenance, the
thymidine, Cotsarelis et al., found slow cycling or LRCs limbal epithelial cells are thought to act as a barrier to the
located in the limbal basal region of the mouse cornea and conjunctival epithelial cells [27]. Ambati et al., has recently
postulated that 10% of limbal basal cells were stem cells. shown that soluble vascular endothelial growth factor
This population of limbal basal cells phenotypically appear receptor 1 (sFlt1) plays an important role in corneal
to be more primitive as they are small and round [1]. avascularity [28]. Further to this, expression of sFlt1 was
The limbal basal region has areas lacking in differenti- found in the corneal epithelium of normal individuals with
ation markers. For example, the 64kDa cytokeratin 3 (CK3) less seen in vascularised patients [29]. When the limbus is
was found in all layers of the corneal epithelium and the non functional, the conjunctiva can invade the corneal
suprabasal layers of the limbal epithelium, however it was epithelium leading to chronic inflammation, neovascular-
absent from the limbal basal cells and the adjacent isation and corneal opacity. This phenomenon is known as
conjunctiva [2]. A similar pattern was found with the limbal stem cell deficiency and can be attributed to both
corneal specific 55kDa protein, cytokeratin 12 (CK12) [16]. hereditary and acquired conditions. Further clinical evidence
Furthermore, there is a lack of markers such as connexin 43 suggesting the location of LESC was shown by Kenyon and
[17] and involucrin [18], both associated with cells destined Tseng, where they transplanted two limbal explants taken
for differentiation. Interestingly, the limbal basal region from the contralateral healthy eye of patients onto the other
expresses progenitor cell markers such as the transcription damaged eye. This resulted in re-epithelisation of the cornea
factor p63 [9], especially the ΔNp63α isoform [19], the and regression of persistent epithelial defects and neo-
ATP-binding cassette transporter Bcrp1/ABCG2 and more vascularisation [30]. This initial work has lead to the use
recently N-cadherin [20]. of expanded LESC grown on amniotic membrane [31] and
Stem Cell Rev (2008) 4:159–168 161

more recently the use of autologous mucosal epithelial cell are heterogeneous, containing limbal fibroblasts which have
grafts [32]. been found to preferentially express secreted protein acidic
and rich in cysteine (SPARC) when compared to corneal
fibroblasts, which may contribute to LESC adhesion [47].
The LESC Niche Furthermore, Nakamura et al. [48], identified a population of
bone marrow-derived cells located in the limbal stroma
The surrounding microenvironment or niche of a stem cell, following transplantation of GFP labelled bone marrow cells
which consists of cellular and extracellular components, is into nude mice. This study suggested these cells are able to
hypothesised to prevent them from differentiating and thus migrate into the limbal stroma, however functionality was
determines their fate [33, 34]. Once a stem cell divides not discussed and should be explored. The interaction
asymmetrically and leaves its niche it enters a differentia- between the limbal stromal cells and LESC is not fully
tion pathway under the influence of different environmental understood and needs further investigation.
stimuli. Interestingly, the mechanisms by which this occurs This specialised microenvironment also provides protec-
still remains unclear. This theory is in keeping with the tion. Firstly, the LESC are located in the basal layer, therefore
LESC niche as it differs from the remaining corneal stroma harmful agents must penetrate the full thickness of the corneal
both anatomically and functionally. epithelium prior to reaching the stem cells. The deep
The LESC niche is thought to be located in the area of the undulations of the of Vogt at the limbus provide LESC with
Palisades of Vogt (PV) which undulate providing increased a protective environment away from the external surroundings
surface area. They are highly pigmented, due to presence of and also protect against shearing forces [49] along with the
melanocytes [12, 35] and infiltrated with Langerhans cells non-uniform junctions between limbal epithelium and
[36] and T-lymphocytes [37]. The melanin pigmentation stroma. Recently, Shortt et al. [39] has found limbal
shields cells from damaging ultraviolet light and the resultant epithelial crypts and focal stromal projections (FSP) are
generation of reactive oxygen species [38]. When comparing predominantly located at the superior and inferior aspects of
the LESC niche to the cornea, the basement membrane the cornea, which offers further protection by the eyelid.
contains papillae of stroma which project upwards [39] and
is also fenestrated [40] suggesting close interaction with
surrounding stromal cells. The limbal and corneal basement Limbal Epithelial Stem Cell Markers
membrane components also differ, with the limbal region
containing laminin-1,5 and α2β2 chains which are not found Much effort has been made to identify potential molecular
in the cornea. Furthermore, type IV collagen α1, α2 and α5 markers to distinguish LESC from TAC located in the limbal
chains are found in the limbal region whereas α3 and α5 are and corneal basal regions. As discussed below, some
located in the cornea [41, 42]. A more recent study by progress has been made using varied approaches, however
Schlötzer-Schrehardt et al., found patchy immunolocalisation there remains controversy with no one molecule being
of laminin γ3 chain, BM40/SPARC and tenancin C, which definitive. Markers can be divided into two subgroups, both
was also found to co-localise with ABCG2/p63/K19-positive negative and positive. Negative markers are absent mole-
cell clusters, suggesting they may be involved in retaining cules from the limbal basal cells and include the cytokeratins
cell stemness [43]. CK3 and CK12 [2] the gap junction protein Cx43. With
It has also been suggested the LESC niche basement positive markers, such as p63 and ABCG2, being located in
membrane may sequester and therefore modulate growth the LESC themselves.
factors and cytokines involved in LESC function allowing Pellegrini et al. [9], initially proposed the transcriptional
for efficient and precise cellular control [44]. Interestingly, factor p63 as a putative LESC marker, as it was found to
it has been recently found that hypoxic conditions in vitro distinguish human keratinocyte stem cells from their TA
produce larger cell colonies and a decrease in differentia- progeny and was therefore suggested to be a epidermal
tion markers, suggesting low oxygen levels induces stem cell marker. Through clonal analysis and western
selective proliferation of undifferentiated cells [45]. Al- blotting, they demonstrated that p63 was expressed in
though the cornea is exposed to atmospheric oxygen, the limbal holoclones with little or none being seen in
LESC niche lies beneath a number of cell layers and meroclones and paraclones. Immunohistochemically, p63
therefore the environment may have lower oxygen levels, was located to limbal basal epithelium, supporting its role
which help maintain the stemness of the cells. as a LESC marker. p63 has also been localised to basal cells
Interestingly, beneath the basement membrane the limbal of the peripheral and central cornea in humans [18, 50] and
niche is vascularised and highly innervated [46] compared to in rats [51] thus ruling it out as a exclusive LESC marker.
the avascular cornea thus potentially providing extra nutrients However, when combining high p63 expression with
and growth factors to LESC. The cells residing in the stroma nuclear/cytoplasmic ratio a small population of cells has
162 Stem Cell Rev (2008) 4:159–168

been identified and suggested to represent LESC [52]. integrin β1 ligand, collagen IV, display many LESC
More recently, the isoform, ΔNp63α has been proposed to properties [58]. Although it is found to be expressed in
be more specific and therefore has been suggested to be a limbal and corneal cells, suggesting it is expressed by more
stem cell marker [19]. differentiated cells, limbal basal cells are said to be integrin
Many types of organ-specific stem cells have been β1 bright.
recently shown to exhibit a side population (SP) phenotype N-cadherin mediates cell–cell adhesion and has been
based on their ability to efflux Hoechst 33342 dye through suggested to play an important role in the maintenance of
the ATP-binding cassette transporter Bcrp1/ABCG2 and haemopoietic stem cells, by facilitating adhesion to osteo-
therefore it has been proposed to be a universal marker for blasts in the bone marrow niche [59, 60]. With this in mind,
stem cells [53]. Flow cytometric and real time RT PCR Hayashi et al, explored the possibility of role for N-
analysis of limbal basal cells has shown these cells to cadherin in LESC maintenance. They found expression of
express this same SP and was therefore suggested as a N-cadherin in a subpopulation of limbal epithelial basal
putative LESC marker [10]. In support, the protein has been cells, suggesting they were putative stem cells. Further to
immunolocalised to the cell membrane and cytoplasm of a this, they also found expression in adjacent melanocytes
population of limbal basal cells and a few suprabasal cells implying N-cadherin plays an important role in interactions
[18] with ABCG2 positive cells producing higher colony between LESC and their corresponding niche cells [20].
forming efficiencies than their negative counterparts [54]. As the limbal epithelium is derived from the neural
Furthermore, our laboratory has localised ABCG2 to the ectoderm a number of neural stem cell markers have been
outer edge of holoclones where it is thought that the stem suggested as LESC markers. Nerve growth factor (NGF) has
cells reside (Fig. 2). Indeed, these studies suggest it is an been associated with the proliferation and differentiation of
ideal candidate as a LESC marker, however further human corneal epithelium with stimulation of its receptor,
clarification is needed. TrkA, being thought to promote cell survival [61]. This high
Integrins such as α9 have been suggested as a possible affinity receptor is expressed in both corneal and limbal basal
marker as it has been localised to small clusters of cells in cells compared with the low affinity NGF receptor, p75NTR
the limbal basal epithelium [55]. However, murine studies which is localised to suprabasal limbal and entire corneal
have demonstrated an upregulation of α9 in wounded epithelium. As TrkA lacks specificity it cannot be considered
corneas, suggesting it is expressed in TAC’s as opposed to a LESC marker, however p75NTR could be looked upon as a
LESC [56]. One other integrin β1 has been associated with marker of differentiation. Recent in depth immunological
LESC, it was originally suggested to be a keratinocyte studies of neurotrophic factors and their receptors in the
marker [57]. Furthermore cells that rapidly adhere to the human has found NGF, Glial cell-derived neurotrophic factor

Fig. 2 Characteristics of limbal


epithelial stem cells in vitro.
When seeded at low density,
limbal epithelial stem cells pro-
duce large holoclone colonies
(a, stained with haematoxylin
and b, phase contrast). The cells
at the outer edge of holoclones
express ABCG2 (c, green),
ΔNp63α (d, red) a merge of the
two markers is shown in e with
DAPI staining of the nuclei
Stem Cell Rev (2008) 4:159–168 163

(GDNF) and their corresponding receptors TrkA and GDNF the least differentiated and therefore may be more stem like
family receptor alpha (GFRα)-1 are exclusively expressed in in nature. Cross talk between cells is facilitated by gap
the limbus [62]. junctions with connexin 43 and connexin 50 being localised
Intermediate filaments such as vimentin, keratin 15 to the corneal epithelium [66]. Cx 43 is expressed by corneal
(K15) and keratin 19 (K19) have been localised to basal basal cells except that of the limbus, implying it is utilised by
cells of the limbus [63]. more early TACs. The lack of intracellular communication
Notch 1 is a ligand-activated transmembrane receptor, has been suggested to help maintain stem cells and their
which has been shown to maintain progenitor cells in other niche [17] and protects the cells from damage affecting its
systems in an undifferentiated state. Antibody staining adjacent neighbouring cells [51]. Involucrin is produced by
demonstrated cell clusters in the Palisaids of Vogt with some all cells within stratified squamous epithelia apart from basal
co-localisation with ABCG2, thus suggesting it may be a cells [67] this also being true for the corneal epithelium [18].
LESC marker [64]. However, others have suggested it plays Furthermore, larger cells from human corneal epithelial
a role in differentiation, proliferation and corneal repair. cultures display increased expression of involucrin, this
Using Notch 1 deficient mice, Vauclair et al, demonstrated along with immunological staining suggests it is also a
Notch 1 signalling is required for cell fate maintenance marker of differentiation.
during corneal epithelial wound healing linking this to In depth immunohistochemical panels have been
regulation of vitamin A metabolism [8]. Furthermore, Ma assessed in attempt to identify informative markers. These
et al. [65], found Notch 1 and other Notch family members latest findings are summarised in Table 1 and suggest when
and their receptors to be expressed throughout the cornea and comparing limbal basal epithelial cells to corneal basal
suggested a role in differentiation. cells, they are positive for K19, ABCG2, vimentin, KGF-R,
Opposed to the identification of a LESC marker is the metallothionein and integrin α9. Furthermore they are
existence of markers of corneal epithelial differentiation. The found to be negative for CK3 and CK12, Cx43, E-cadherin,
corneal specific differentiation markers K3 and K12 are involucrin and integrins α2, α6 and β4 [68]. The majority
expressed throughout the corneal epithelium but are absent of LESC putative markers are expressed in most limbal
from the limbal basal cells, thus indicating that these cells are basal cells and some suprabasal cells, therefore it cannot be

Table 1 Localisation of puta-


tive stem cell markers of the Limbal epithelium Corneal epithelium
corneal epithelium
Basal Suprabasal Basal Suprabasal

Positive markers
p63 +++ +/− +/− −
Δp63α ++ − − −
ABCG2 +++ +/− − −
Integrin α9 +++ +/− − −
NGF R (TrkA) +++ ++ +++ ++
α-enolase +++ + ++ +
Vimentin +++ +/− − −
Keratin 19 ++ − − −
Integrin β1 +++ ++ ++ +
N-Cadherin ++ − − −
GDNF +++ ++ − −
GFRα-1 +++ ++ − −
Keratin 15 ++ ++ − −
KGF-R +/− − − −
Negative markers
CK3/CK12 − +++ +++ +++
Cx 43 − +++ + +++
Cx 50 − +++ − +++
Involucrin − +++ + +++
NGFR(p75NTR) − +++ +++ +++
Adapted from [68] and [51] E-cadherin +/− +++ +++ +++
− No staining, +/− very weak Integrin α2 − or +++ ++ +++ +++
staining, + weak staining, ++ Integrin α6 − or +++ ++ +++ +++
moderate staining, +++ strong Integrin β4 − or +++ ++ +++ +++
staining
164 Stem Cell Rev (2008) 4:159–168

ruled out that these markers also present in early TAC. The underlying process of these abnormalities is poorly
Currently the most promising candidate markers are understood and is thought to be due to stem cell failure [78–
ABCG2, Notch-1 and N-cadherin. ABCG2 and Notch 1 80] however, it has been proposed that it may be due to a
both identify small clusters of limbal basal cells, with N- deficiency in the stem cell niche and adjacent corneal stroma
cadherin also identifying adjacent melanocytes. [81] and more recently a downregulation of PAX6 has been
linked to abnormal epidermal differentiation of cornea
epithelial cells [82].
LESC Deficiency An important factor leading to progressive loss of vision
is aniridic-related keratopathy (ARK) [78, 83] which occurs
Partial or full LESC deficiency occurs as a result of damage in 90% of patients. Initially the cornea of patients appears
or disease to the LESC population, which leads to normal during childhood [79, 84]. Changes occur in
deleterious effects on corneal wound healing and surface patients in their early teenage years, with the disease
integrity [50, 69]. Deficiency can arise from injuries manifesting as a thickened irregular peripheral epithelium.
including chemical or thermal burns and through diseases This is followed by superficial neovascularisation and if left
such as Stevens Johnson syndrome and aniridia. Character- untreated it may result in subepithelial fibrosis and stromal
istics of LESC deficiency include ingrowth of the conjunc- scarring (Fig. 3). Furthermore patients develop recurrent
tival epithelium, vascularisation, chronic inflammation, erosions, ulcerations, chronic pain and eventual blindness
recurrent erosions and ulceration, basement membrane loss [85]. Histologically, stromal neovascularisation and infil-
and ingrowth of fibrotic tissue. This leads to functional tration of inflammatory cells is seen with the destruction of
impairment and eventual visual loss [30, 70, 71]. Long term Bowman’s layer. Additionally, the presence of goblet and
restoration of visual function requires renewal of the conjunctival cells is seen on the corneal surface [83].
corneal epithelium, through replacement of the stem cell Traditionally, these clinical and histological manifestations
population by grafting limbal auto- or allografts [30]. has lead to the consensus that LESC deficiency is largely
Traditionally, treatment of LESC deficient patients has responsible for corneal abnormalities in aniridia [83, 86].
been achieved by grafting viable limbal tissue, from either the
fellow healthy eye or a donor eye, thus replacing the LESC
population [72]. A variety of limbal tissue transplantation
methods have been used, including cadaveric keratolimbal
allograft (KLAL), live or living related conjunctival limbal
allograft (Ir-CLAL) and limbal autograft. Each procedure
carries a risk of complication such as damage to healthy eye
with removal of autologous grafts and complications due to
long term immunosuppression with allogenic tissue, there-
fore new strategies have been developed. Once such
procedure is transplantation of cultured LESC grown on a
suitable substrate such as amnionic membrane or fibrin, with
or without growth arrested 3T3 fibroblast feeder layers [73–
77]. The mechanisms behind this method are not clearly
defined however, initial success rates are promising.

PAX6 Insufficiency and LESC Deficiency

Aniridia

One of the causes of blindness in children with aniridia is due


to progressive ocular surface failure. The disease is caused by
PAX6 heterozygosity, which leads to a pan-ocular, bilateral
condition most prominently characterised by iris hypoplasia,
which varies from a relatively normal iris to the complete lack Fig. 3 Aniridia. An aniridic human eye (a) showing neovascularisa-
tion, inflammation and opacity of the cornea. WT mice have a normal
of an iris. Aniridia is often associated with cataracts, corneal complement of Pax6 gene expression and normal eyes (b). The Pax6
vascularisation and glaucoma, with a significant number of heterozygous sey small eyed mouse with LESC failure is a model of
cases of visual morbidity being due to corneal abnormalities. human aniridia (c)
Stem Cell Rev (2008) 4:159–168 165

As a LESC marker has yet to be definitively identified, a corneal epithelial cells [99]. It has been found that CK12
true demonstration of LESC deficiency cannot be assumed. expression is Pax6 dependant [99] with CK12 deficient mice
Furthermore treatment for these patients involving replace- producing a fragile superficial corneal epithelium that often
ment of LESC, either by keratolimbal allografts or more detaches [100]. This phenotype is also seen in the Pax6+/−
recently ex vivo expanded LESC grafts, provides a better mice [72] and when comparing them to Pax6+/+mice, there
outcome than corneal transplants [77, 85, 87], this being is a decrease in CK12 staining. Further to this, aniridic
consistent with LESC deficiency. However, patients who patients also have a decrease in CK12 staining [80]. This
receive both limbal and corneal tissue seem to have the suggests the corneal fragility seen in both Pax6+/− mice and
better outcome, suggesting an abnormality with corneal aniridic patients is due poor differentiation as a consequence
tissue and not just the limbus. This may be a downstream of lowered levels of CK12, being a downstream of altered
effect of LESC deficiency or that low levels of Pax6 has a Pax6 expression. A recent human study has suggested that
generalised effect on the entire cornea. Alternatively, ARK loss of PAX6 and resultant down regulation of CK12 seen in
may be a consequence of an abnormal corneal epithelial/ squamous metaplasia results in abnormal epidermal differ-
stromal healing response as there is insufficient evidence to entiation as evidenced by expression of CK10 and filaggrin
indicate that the proliferative potential of LESC is impaired both known epidermal proteins. Furthermore, due to local-
[81, 88]. Recently, studies looking at the regulation of isation of CK10 expression to the suprabasal cells in the
genes downstream of Pax6 in the Pax6 mutant mouse, limbal and peripheral cornea suggests PAX6 controls LESC
suggests the pathogenesis of ARK is due a number of plasticity and a loss leads to transdifferentiation in squamous
mechanisms and not solely due to LESC deficiency [81]. metaplasia and could also indicate a role for the limbal stem
Further studies are needed to elucidate the exact mecha- cell niche [101].
nisms of ARK progression to allow the use of appropriate Sonic hedgehog, Wnt/β-catenin, TGF-β and Notch
treatments. signalling pathways have all being implicated in niche
Heterozygous Pax6+/− mice or small eye (sey) mice, control of stem cells, however the LESC and their niche is
provide an excellent model for aniridia and the progressive still relatively unchartered. It has recently been shown that
nature of associated corneal abnormalities [89, 90]. As the mice lacking in expression of Dkk2, a Wnt pathway
name suggests, mice with semidominant mutations develop inhibitor, display epidermal differentiation on the ocular
small eyes and other ocular deformities (Fig. 3.). Homo- surface. The lack of Dkk2, leads to increased Wnt/β-
zygotes generate an ultimately lethal phenotype with no catenin signalling in the limbal stroma, demonstrating the
eyes and nasal primordial [91, 92]. A number of sey mice importance of limbal niche control over LESC differentia-
arose independently all of which are semidominant and by tion during development. Pax6 expression is lost in the
examining comparative mapping studies and phenotypic corneal epithelial cells of these mice, suggesting it is
similarities to aniridia, it was suggested to be the mouse downstream of Dkk2 [102]. Furthermore a small population
homologue of the human disease [92]. This research led to of progenitor corneal fibroblast or keratocytes has been
the discovery that the Pax6 gene was responsible for the identified as being Pax6 positive [103]. Down regulation of
Sey phenotype and suggested that it was also responsible Pax6 in these cells could lead to altered LESC control.
for the human disease, aniridia [91]. Deficiencies in PAX6 resulting in impaired corneal epithe-
lial function and eventual LESC failure may be due to
The Pax6 Gene altered niche development.

The PAX6 gene is a hierarchical transcriptional factor Summary


regulating a multitude of genes involved in eye develop-
ment and adult homeostasis. The PAX6 protein has been Limbal epithelial stem cell health is imperative to normal
found to be expressed in the adult eye, cerebellum and corneal function with deficiencies leading to conjunctival-
pancreas suggesting it has an important role in maintenance isation, neovascularisation and eventual visual loss due to
and remodelling of adult tissues [93–96]. PAX6/Pax6 is corneal opacity. One such disease that has been considered
involved in a number of regulatory roles in the adult cornea to be a consequence of LESC deficiency is aniridia, which
and is essential for the expression of Cytokeratin 12, is caused by mutations in the PAX6 gene. A better
gelatinase B (MMP-9) and cell adhesion molecules (CAM) understanding of PAX6 regulation of LESCs through the
[89, 96, 97] and more recently it has been suggested that use of animal models has helped to gain insight into LESC
knockdown of Pax6 expression promotes EGF-induced cell characteristics and function. However, LESC control is still
proliferation [98]. relatively uncharted and therefore a better understanding of
Cytokeratins (CK) are structural proteins expressed by regulation will improve disease prevention, progression and
epithelial cells, with CK12 being a specific for differentiated treatment in the future.
166 Stem Cell Rev (2008) 4:159–168

Acknowledgements This work was supported by the ERANDA lack connexins and metabolite transfer capacity. Differentiation,
Foundation (GAS), the Special Trustees of Moorfields Eye Hospital 61, 251–260.
and the NIHR BMRC for Ophthalmology (JTD). Thank you to 18. Chen, Z., de Paiva, C. S., Luo, L., Kretzer, F. L., Pflugfelder, S. C.,
Mr. Alex Shortt for the clinical data. & Li, D. Q. (2004). Characterization of putative stem cell phenotype
in human limbal epithelia. Stem Cells, 22, 355–366.
19. Di Iorio, E., Barbaro, V., Ruzza, A., Ponzin, D., Pellegrini, G., &
de Luca, M. (2005). Isoforms of DeltaNp63 and the migration of
References
ocular limbal cells in human corneal regeneration. Proceedings
of the National Academy of Sciences of the United States of
1. Romano, A. C., Espana, E. M., Yoo, S. H., Budak, M. T., America, 102, 9523–9528.
Wolosin, J. M., & Tseng, S. C. (2003). Different cell sized in 20. Hayashi, R., Yamato, M., Sugiyama, H., et al. (2007). N-
human limbal and central corneal basal epithelia measured by Cadherin is expressed by putative stem/progenitor cells and
confocal microscopy and flow cytometry. Investigative Ophthal- melanocytes in the human limbal epithelial stem cell niche. Stem
mology & Visual Science, 44, 5125–5129. Cells, 25, 289–296.
2. Schermer, A., Galvin, S., & Sun, T. T. (1986). Differentiation- 21. Lavker, R. M., Dong, G., Cheng, S. Z., Kudoh, K., Cotsarelis, G., &
related expression of a major 64K corneal keratin in vivo and in Sun, T. T. (1991). Relative proliferative rates of limbal and
culture suggests limbal location of corneal epithelial stem cells. corneal epithelia. Implications of corneal epithelial migration,
Journal of Cell Biology, 103, 49–62. circadian rhythm, and suprabasally located DNA-synthesizing
3. Kurpakus, M. A., Stock, E. L., & Jones, J. C. (1990). Expression keratinocytes. Investigative Ophthalmology & Visual Science, 32,
of the 55-kD/64-kD corneal keratins in ocular surface epitheli- 1864–1875.
um. Investigative Ophthalmology & Visual Science, 31, 22. Ebato, B., Friend, J., & Thoft, R. A. (1987). Comparison of
448–456. central and peripheral human corneal epithelium in tissue
4. Barrandon, Y., & Green, H. (1987). Three clonal types of culture. Investigative Ophthalmology & Visual Science, 28,
keratinocyte with different capacities for multiplication. Proceed- 1450–1456.
ings of the National Academy of Sciences of the United States of 23. Ebato, B., Friend, J., & Thoft, R. A. (1988). Comparison of
America, 84, 2302–2306. limbal and peripheral human corneal epithelium in tissue culture.
5. Cotsarelis, G., Cheng, G., Dong, G., Sun, T. T., & Lavker, R. M. Investigative Ophthalmology & Visual Science, 29, 1533–1537.
(1989). Existence of slow-cycling limbal epithelial basal cells 24. Pellegrini, G., Golisano, O., Paterna, P., et al. (1999). Location and
that can be preferentially stimulated to proliferate: Implications clonal analysis of stem cells and their differentiated progeny in the
on epithelial stem cells. Cell, 57, 201–209. human ocular surface. Journal of Cell Biology, 145, 769–782.
6. Lavker, R. M., & Sun, T. T. (2003). Epithelial stem cells: The 25. Kruse, F. E., & Tseng, S. C. (1993). A tumor promoter-resistant
eye provides a vision. Eye, 17, 937–942. subpopulation of progenitor cells is larger in limbal epithelium
7. Lehrer, M. S., Sun, T. T., & Lavker, R. M. (1998). Strategies of than in corneal epithelium. Investigative Ophthalmology &
epithelial repair: Modulation o stem cell and transit amplifying Visual Science, 34, 2501–2511.
cell proliferation. Journal of Cell Science, 111, 2867–2875. 26. Lavker, R. M., Wei, Z. G., & Sun, T. T. (1998). Phorbol ester
8. Vauclair, S., Majo, F., Durham, A. D., Ghyselinck, N. B., preferentially stimulates mouse fornical conjunctival and limbal
Barrandon, Y., & Radtke, F. (2007). Corneal epithelial cell fate is epithelial cells to proliferate in vivo. Investigative Ophthalmol-
maintained during repair by Notch 1 signalling via the regulation ogy & Visual Science, 39, 301–307.
of vitamin A metabolism. Developmental Cell, 13, 12. 27. Tseng, S. C. (1989). Concept and application of limbal stem
9. Pellegrini, G., Dellambra, E., Golisano, O., et al. (2001). p63 cells. Eye, 3(Pt 2), 141–157.
identifies keratinocyte stem cells. Proceedings of the National 28. Ambati, B. K., Nozaki, M., Singh, N., et al. (2006). Corneal
Academy of Sciences of the United States of America, 98, avascularity is due to soluble VEGF receptor-1. Nature, 443,
3156–3161. 993–997.
10. Watanabe, K., Nishida, K., Yamato, M., et al. (2004). Human limbal 29. Ambati, B. K., Patterson, E., Jani, P., et al. (2007). Soluble
epithelium contains side population cells expressing the ATP- vascular endothelial growth factor receptor-1 contributes to the
binding cassette transporter ABCG2. FEBS Letters, 565, 6–10. corneal antiangiogenic barrier. British Journal of Ophthalmol-
11. Mann, I. (1944). A study of epithelial regeneration in the living ogy, 91, 505–508.
eye. British Journal of Ophthalmology, 28, 26–40. 30. Kenyon, K. R., & Tseng, S. C. (1989). Limbal autograft
12. Davanger, M., & Evenson, A. (1971). Role of the pericorneal transplantation for ocular surface disorders. Ophthalmology, 96,
structure in renewal of corneal epithelium. Nature, 229, 560–561. 709–722.
13. Huang, A. J., & Tseng, S. C. (1991). Corneal epithelial wound 31. Tsai, R. J.-F., Li, L.-M., & Chen, J.-K. (2000). Reconstruction of
healing in the absence of limbal epithelium. Investigative damaged corneas by transplantation of autologous limbal
Ophthalmology & Visual Science, 32, 96–105. epithelial cells. New England Journal of Medicine, 343, 86–93.
14. Kinoshita, S., Friend, J., & Thoft, R. A. (1981). Sex chromatin of 32. Nakamura, M., Endo, K.-I., Cooper, L. J., et al. (2003). The
donor corneal epithelium in rabbits. Investigative Ophthalmology successful culture and autologous transplantation of rabbit oral
& Visual Science, 21, 434–441. mucosal epithelial cells on amniotic membrane. Investigative
15. Bickenbach, J. R. (1986). Identification and behavior of label- Ophthalmology & Visual Science, 44, 106–116.
retaining cells in oral mucosa and skin. Journal of Dental 33. Schofield, R. (1983). The stem cell system. Biomedicine &
Research, 60(Spec No C), 1611–1620. Pharmacotherapy, 37, 375–380.
16. Chaloin-Dufau, C., Sun, T. T., & Dhouailly, D. (1990). 34. Watt, F. M., & Hogan, B. L. (2000). Out of Eden: stem cells and
Appearance of the keratin pair K3/K12 during embryonic and their niches. Science, 287, 1427–1430.
adult corneal epithelial differentiation in the chick and in the 35. Higa, K., Shimmura, S., Miyashita, H., Shimazaki, J., &
rabbit. Cell Differentiation and Development, 32, 97–108. Tsubota, K. (2005). Melanocytes in the corneal limbus interact
17. Matic, M., Petrov, I. N., Chen, S., Wang, C., Dimitrijevich, S. D., with K19-positive basal epithelial cells. Experimental Eye
& Wolosin, J. M. (1997). Stem cells of the corneal epithelium Research, 81, 218–223.
Stem Cell Rev (2008) 4:159–168 167

36. Baum, J. L. (1970). Melanocyte and Langerhans cell population 54. de Paiva, C. S., Chen, Z., Corrales, R. M., Pflugfelder, S. C., &
of the cornea and limbus in the albino animal. American Journal Li, D. Q. (2005). ABCG2 transporter identifies a population of
of Ophthalmology, 69, 669–676. clonogenic human limbal epithelial cells. Stem Cells, 23, 63–73.
37. Vantrappen, L., Geboes, K., Missotten, L., Maudgal, P. C., & 55. Stepp, M. A., Zhu, L., Sheppard, D., & Cranfill, R. L. (1995).
Desmet, V. (1985). Lymphocytes and Langerhans cells in the Localized distribution of alpha 9 integrin in the cornea and
normal human cornea. Investigative Ophthalmology & Visual changes in expression during corneal epithelial cell differentia-
Science, 26, 220–225. tion. Journal of Histochemistry Cytochemistry, 43, 353–362.
38. Shimmura, S., & Tsubota, K. (1997). Ultraviolet B-induced 56. Stepp, M. A., & Zhu, L. (1997). Upregulation of alpha 9 integrin
mitochondrial dysfunction is associated with decreased cell and tenascin during epithelial regeneration after debridement in the
detachment of corneal epithelial cells in vitro. Investigative cornea. Journal of Histochemistry Cytochemistry, 45, 189–201.
Ophthalmology & Visual Science, 38, 620–626. 57. Jones, P. H., & Watt, F. M. (1993). Separation of human epidermal
39. Shortt, A. J., Secker, G. A., Munro, P. M., Khaw, P. T., Tuft, S. J., stem cells from transit amplifying cells on the basis of differences
& Daniels, J. T. (2007). Characterisation of the limbal epithelial in integrin function and expression. Cell, 73, 713–724.
stem cell niche: novel imaging techniques permit in-vivo 58. Li, D. Q., Chen, Z., Song, X. J., de Paiva, C. S., Kim, H. S., &
observation and targeted biopsy of limbal epithelial stem cells. Pflugfelder, S. C. (2005). Partial enrichment of a population of
Stem Cells, 5, 1402–1409. human limbal epithelial cells with putative stem cell properties
40. Dua, H. S., Shanmuganathan, V. A., Powell-Richards, A. O., based on collagen type IV adhesiveness. Experimental Eye
Tighe, P. J., & Joseph, A. (2005). Limbal epithelial crypts: a Research, 80, 581–590.
novel anatomical structure and a putative limbal stem cell niche. 59. Zhang, J., Niu, C., Ye, L., et al. (2003). Identification of the
British Journal of Ophthalmology, 89, 529–532. haematopoietic stem cell niche and control of the niche size.
41. Ljubimov, A. V., Burgeson, R. E., Butkowski, R. J., Micheal, A. F., Nature, 425, 836–841.
Sun, T. T., & Kenney, M. C. (1995). Human corneal basement 60. Calvi, L. M., Adams, G. B., Weibrecht, K. W., et al. (2003).
membrane heterogeneity: topographical differences in the expres- Osteoblastic cells regulate the haematopoietic stem cell niche.
sion of type IV collagen and laminin isoforms. Laboratory Nature, 425, 841–846.
Investigation, 72, 461–473. 61. Lambiase, A., Bonini, S., Micera, A., Rama, P., & Aloe, L.
42. Tuori, A., Uusitalo, H., et al. (1996). The immunohistochemical (1998). Expression of nerve growth factor receptors on the
composition of the human corneal basement membrane. Cornea, ocular surface in healthy subjects and during manifestation of
15, 286–294. inflammatory diseases. Investigative Ophthalmology & Visual
43. Schlotzer-Schrehardt, U., Dietrich, T., Saito, K., et al. (2007). Science, 39, 1272–1275.
Characterization of extracellular matrix components in the limbal 62. Qi, H., Li, D. Q., Shine, H. D., et al. (2008). Nerve growth factor
epithelial stem cell compartment. Experimental Eye Research, and its receptor TrkA serve as potential markers for human
85, 845–860. corneal epithelial progenitor cells. Experimental Eye Research,
44. Klenkler, B., & Sheardown, H. (2004). Growth factors in the 86, 34–40.
anterior segment: Role in tissue maintenance, wound healing and 63. Yoshida, S., Shimmura, S., Kawakita, T., et al. (2006).
ocular pathology. Experimental Eye Research, 79, 677–688. Cytokeratin 15 can be used to identify the limbal phenotype in
45. Miyashita, H., Higa, K., Kato, N., et al. (2007). Hypoxia enhances normal and diseased ocular surfaces. Investigative Ophthalmol-
the expansion of human limbal epithelial progenitor cells in vitro. ogy & Visual Science, 47, 4780–4786.
Investigative Ophthalmology & Visual Science, 48, 3586–3593. 64. Thomas, P. B., Liu, Y. H., Zhuang, F. F., et al. (2007).
46. Lawrenson, J. G., & Ruskell, G. L. (1991). The structure of Identification of Notch-1 expression in the limbal basal
corpuscular nerve endings in the limbal conjunctiva of the epithelium. Molecular Vision, 13, 337–344.
human eye. Journal of Anatomy, 177, 75–84. 65. Ma, A., Boulton, M., Zhao, B., Connon, C., Cai, J., & Albon, J.
47. Shimmura, S., Miyashita, H., Higa, K., Yoshida, S., Shimazaki, J., (2007). A role for notch signaling in human corneal epithelial
& Tsubota, K. (2006). Proteomic analysis of soluble factors cell differentiation and proliferation. Investigative Ophthalmol-
secreted by limbal fibroblasts. Molecular Vision, 12, 478–484. ogy & Visual Science, 48, 3576–3585.
48. Nakamura, T., Ishikawa, F., Sonoda, K. H., et al. (2005). 66. Dong, Y., Roos, M., Gruijters, T., et al. (1994). Differential
Characterization and distribution of bone marrow-derived cells expression of two gap junctions proteins in corneal epithelium.
in mouse cornea. Investigative Ophthalmology & Visual Science, European Journal of Cell Biology, 64, 95–100.
46, 497–503. 67. Watt, F. M., & Green, H. (1981). Involucrin synthesis is
49. Gipson, I. K. (1989). The epithelial basement membrane zone of correlated with cell size in human epidermal cultures. Journal
the limbus. Eye, 3, 132–140. of Cell Biology, 90, 738–742.
50. Dua, H. S., Joseph, A., Shanmuganathan, V. A., & Jones, R. E. 68. Schlotzer-Schrehardt, U., & Kruse, F. E. (2005). Identification
(2003). Stem cell differentiation and the effects of deficiency. and characterisation of limbal stem cells. Experimental Eye
Eye, 17, 877–885. Research, 81, 247–264.
51. Chee, K. Y., Kicic, A., & Wiffen, S. J. (2006). Limbal stem cells: 69. Chen, J. J., & Tseng, S. C. (1991). Abnormal corneal epithelial
the search for a marker. Clinical & Experimental Ophthalmol- wound healing in partial-thickness removal of limbal epithelium.
ogy, 34, 64–73. Investigative Ophthalmology & Visual Science, 32, 2219–33.
52. Arpitha, P., Prajna, N. V., Srinivasan, M., & Muthukkaruppan, V. 70. Puangsricharern, V., & Tseng, S. C. (1995). Cytologic evidence
(2005). High expression of p63 combined with a large N/C ratio of corneal diseases with limbal stem cell deficiency. Ophthal-
defines a subset of human limbal epithelial cells: implications on mology, 102, 1476–1485.
epithelial stem cells. Investigative Ophthalmology & Visual 71. Holland, E. J., & Schwartz, G. S. (1996). The evolution of
Science, 46, 3631–3636. epithelial transplantation for severe ocular surface disease and a
53. Zhou, S., Schuetz, J. D., Bunting, K. D., et al. (2001). The ABC proposed classification system. Cornea, 15, 549–556.
transporter Bcrp1/ABCG2 is expressed in a wide variety of stem 72. Ramaesh, K., & Dhillon, B. (2003). Ex vivo expansion of
cells and is a molecular determinant of the side-population corneal limbal epithelial/stem cells for corneal surface recon-
phenotype. Natural Medicines, 7, 1028–1034. struction. European Journal of Ophthalmology, 13, 515–524.
168 Stem Cell Rev (2008) 4:159–168

73. Lindberg, K., Brown, M. E., Chaves, H. V., Kenyon, K. R., & 88. Sivak, J. M., Mohan, R., Rinehart, W. B., Xu, P. X., Maas, R. L.,
Rheinwald, J. G. (1993). In vitro propagation of human ocular & Fini, M. E. (2000). Pax-6 expression and activity are induced
surface epithelial cells for transplantation. Investigative Oph- in the reepithelialising cornea and control activity of the
thalmology & Visual Science, 34, 2672–2679. transcriptional promotor for matrix metalloproteinase gelatinase
74. Pellegrini, G., Traverso, C. E., Franzi, A. T., Zingirian, M., B. Developments in Biologicals, 222, 41–54.
Cancedda, R., & De Luca, M. (1997). Long-term restoration of 89. Davis, J., Duncan, M. K., Robison, W. G. J., & Piatigorsky, J.
damaged corneal surfaces with autologous cultivated human (2003). Requirement for Pax6 in corneal morphogenesis: A role
epithelium. The Lancet, 349, 990–993. in adhesion. Journal of Cell Science, 116, 2157–2167.
75. Koizumi, N., Inatomi, T., Suzuki, K., Sotozono, C., & Kinoshita, 90. Ramaesh, T., Collinson, J. M., Ramaesh, K., Kaufman, M. H.,
S. (2001). Cultivated corneal epithelial stem cell transplantation West, J. D., & Dhillon, B. (2003). Corneal abnormalities in Pax6+/−
in ocular surface disorders. Ophthalmology, 108, 1569–1574. small eye mice mimic human aniridia-related keratopathy.
76. Grueterich, M., Espana, E. M., Touhami, A., Ti, S. E., & Investigative Ophthalmology & Visual Science, 44, 1871–1878.
Tseng, S. C. (2002). Phenotypic study of a case with successful 91. Hill, R. E., Favor, J., Hogan, B. L., et al. (1991). Mouse small
transplantation of ex vivo expanded human limbal epithelium eye results from mutations in a paired-like homeobox-containing
for unilateral total limbal stem cell deficiency. Ophthalmology, gene. Nature, 354, 522–525.
109, 1547–1552. 92. Glaser, T., Lane, J., & Housman, D. (1990). A mouse model of the
77. Shortt, A. J., Secker, G. A., Notara, M. D., et al. (2007). aniridia–Wilms tumor deletion syndrome. Science, 250, 823–827.
Transplantation of ex-vivo cultured limbal epithelial stem cells— 93. Ton, C. C., Miwa, H., & Saunders, G. F. (1992). Small eye
a review of current techniques and clinical results. Survey of (Sey): Cloning and characterisation of the murine homolog of the
Ophthalmology, 52, 483–502. human aniridia gene. Genomics, 13, 251–256.
78. Mackman, G., Brightbill, F. S., & Optiz, J. M. (1979). Corneal 94. Turque, N., Plaza, S., Radvanyi, F., Carriere, C., & Saule, S.
changes in aniridia. American Journal of Ophthalmology, 87, (1994). Pax-QNR/Pax6, a paired box- and homeobox-containing
497–502. gene expressed in neurons, is also expressed in pancreatic
79. Nishida, K., Kinoshita, S., Ohashi, Y., Kuwayama, Y., & endocrine cells. Molecular Endocrinology, 8, 929–938.
Yamamoto, S. (1995). Ocular surface abnormalities in aniridia. 95. Stoykova, A., & Gruss, P. (1994). Roles of Pax-6 genes in
American Journal of Ophthalmology, 120, 368–375. developing and adult brain as suggested by expression patterns.
80. Tseng, S. C., & Li, D. Q. (1996). Comparison of protein kinase Journal of Neuroscience, 3, 1395–1412.
C subtype expression between normal and aniridic human ocular 96. Liu, C. Y., Zhu, G., Westerhausen-Larson, A., Converse, R.,
surfaces: implications for limbal cell dysfunction in aniridia. Kao, C. W., & Sun, T. T. (1993). Cornea-specific expression of
Cornea, 15, 168–178. K12 keratin during mouse development. Current Eye Research,
81. Ramaesh, K., Ramaesh, T., Dutton, G. N., & Dhillon, B. (2005). 12, 963–974.
Evolving concepts on the pathogenic mechanisms of aniridia 97. Shiraishim, A., Converse, R. L., Liu, C. Y., Zhou, F., Kao, C. W.,
related keratopathy. International Journal of Biochemistry & & Kao, W. W. (1998). Identification of the cornea-specific
Cell Biology, 37, 547–557. keratin 12 promoter by in vivo particle-mediated gene transfer.
82. Li, W., Chen, Y. T., Hayashida, Y., et al. (2008). Down- Investigative Ophthalmology & Visual Science, 39, 2554–2561.
regulation of Pax6 is associated with abnormal differentiation 98. Li, T., & Lu, L. (2005). Epidermal growth factor-induced
of corneal epithelial cells in severe ocular surface diseases. proliferation requires down-regulation of Pax6 in corneal
Journal of Pathology, 214, 114–122. epithelial cells. Journal of Biological Chemistry, 280, 12988–
83. Margo, C. E. (1983). Congenital aniridia: a histopathologic study 12995.
of the anterior segment in children. Journal of Pediatric 99. Sivak, J. M., & Fini, M. E. (2002). MMPs in the eye: emerging
Ophthalmology and Strabismus, 20, 192–198. roles for matrix metalloproteinases in ocular physiology. Prog-
84. Nelson, L. B., Spaeth, G. L., Nowinski, T. S., Margo, C. E., & ress in Retinal and Eye Research, 21, 1–14.
Jackson, L. (1984). Aniridia. A review. Survey of Ophthalmol- 100. Kao, W. W., Liu, C. Y., Converse, R. L., et al. (1996). Keratin
ogy, 28, 621–642. 12-deficient mice have fragile corneal epithelia. Investigative
85. Holland, E. J., Djalilian, A. R., & Schwartz, G. S. (2003). Ophthalmology & Visual Science, 37, 2572–2584.
Management of aniridic keratopathy with keratolimbal allograft: 101. Li, W., Chen, Y.-T., Hayashida, Y., et al. (2008). Down-regulation
a limbal stem cell transplantation technique. Ophthalmology, of Pax6 is associated with abnormal differentiation of corneal
110, 125–130. epithelial surface diseases. The Journal of Pathology, 214, 9.
86. Dua, H. S., & Azuara-Blanco, A. (2000). Limbal stem cells of 102. Mukhopadhyay, M., Gorivodsky, M., Shtrom, S., et al. (2006).
the corneal epithelium. Survey of Ophthalmology, 44, 415–425. Dkk2 plays an essential role in the corneal fate of the ocular
87. Tiller, A. M., Odenthal, M. T., Verbraak, F. D., & Gortzak- surface epithelium. Development, 133, 2149–2154.
Moorstein, N. (2003). The influence of keratoplasty on visual 103. Funderburgh, M. L., Du, Y., Mann, M. M., SundarRaj, N., &
prognosis in aniridia: a historical review of one large family. Funderburgh, J. L. (2005). PAX6 expression identifies progenitor
Cornea, 22, 105–110. cells for corneal keratocytes. FASEB Journal, 19, 1371–1373.

You might also like