Abstract
Homeostasis of the corneal epithelium is ultimately maintained by stem cells that reside in a specialized microenvironment within the corneal limbus termed palisades of Vogt. This limbal niche nourishes, protects, and regulates quiescence, self-renewal, and fate decision of limbal epithelial stem/progenitor cells (LEPCs) toward corneal epithelial differentiation. This review focuses on our current understanding of the mechanism by which limbal (stromal) niche cells (LNCs) regulate the aforementioned functions of LEPCs. Based on our discovery and characterization of a unique extracellular matrix termed HC-HA/PTX3 (Heavy chain (HC1)-hyaluronan (HA)/pentraxin 3 (PTX3) complex, “-” denotes covalent linkage; “/” denotes non-covalent binding) in the birth tissue, i.e., amniotic membrane and umbilical cord, we put forth a new paradigm that HC-HA/PTX3 serves as a surrogate matrix niche by maintaining the in vivo nuclear Pax6+ neural crest progenitor phenotype to support quiescence and self-renewal but prevent corneal fate decision of LEPCs. This new paradigm helps explain how limbal stem cell deficiency (LSCD) develops in aniridia due to Pax6-haplotype deficiency and further explains why transplantation of HC-HA/PTX3-containing amniotic membrane prevents LSCD in acute chemical burns and Stevens Johnson syndrome, augments the success of autologous LEPCs transplantation in patients suffering from partial or total LSCD, and assists ex vivo expansion (engineering) of a graft containing LEPCs. We thus envisage that this new paradigm based on regenerative matrix HC-HA/PTX3 as a surrogate niche can set a new standard for regenerative medicine in and beyond ophthalmology.
Keywords: amniotic membrane, HC-HA/PTX3, limbus, limbal stem cell deficiency, niche, stem cell
1. Limbal Niche
The corneal epithelium is composed of non-keratinized, stratified squamous epithelial cells held together by tight junctions to protect the cornea from external insult and ensure corneal transparency and clear vision. To maintain corneal epithelial homeostasis and ultimately vision, the corneal epithelium self-renews throughout life from a population of stem cells in the limbal niche (Ang et al., 2004; Cheng et al., 2016; Schermer et al., 1986). Anatomically and clinically, the native limbal niche can be recognized by the pigmented “palisades of Vogt”, which has a vascularized and innervated underlying stroma (Lavker et al., 2004) (Fig. 1A). Palisades are more pronounced in the superior and inferior limbus (Goldberg and Bron, 1982; Townsend, 1991), presumably due to a greater need of limbal epithelial stem cells (LEPCs) to withstand shear stress forces from blinking and progressive attrition of corneal epithelial cells. Other possible explanation suggests the role of eyelids in limbal niche protection from UV radiations may also explain the higher density of LSC in the vertical meridian in humans which is not found in nocturnal mammals such as mice (Grieve et al., 2015). Within this locale, LEPCs reside in the basal epithelial layer (Schermer et al., 1986) between palisade epithelial ridges that extend deep into the stroma beyond the limbal basement membrane to form limbal epithelial crypts (Chen et al., 2011),(Dua et al., 2005),(Kulkarni et al., 2010) (Fig. 1B and 1C). Several studies have found an age-related decline in the number of palisades of Vogt and limbal crypts in humans (Zheng and Xu, 2008),(Notara et al., 2013). This may explain why the basal cell density in the limbal epithelium decreases with age (Niederer et al., 2007),(Tseng, 1989); (Pellegrini et al., 1999), and the epithelial thickness of the limbus and the peripheral cornea gradually deteriorates over time (Yang et al., 2014).
Figure 1: Schematic Representation of Human Limbal Niche.

The human limbal niche (A) is located in a microenvironment termed palisades of Vogt (B, bar = 200 mm), where SCs are within the limbal basal epithelial cells undulated into folds (C) and invaginated into the limbal stroma as crypts (not shown). A subset of mesenchymal cells immediately subjacent to LEPCs functions as LNCs (C). The close interaction between LNCs modulates functions of LEPCs such as quiescence, self-renewal and fate decision (D), of which each process is highlighted by the preferential expression of certain markers specific to the limbus (E). TAC: transient amplifying cells, PMC: post-mitotic cells, TDC: terminally differentiated cells. (Image Taken from (Tseng et al 2016) with modification.)
Compared to corneal epithelial cells, LEPCs are smaller in size with a higher nucleus-to-cytoplasm ratio (Shortt et al., 2007),(Pellegrini et al., 1999),(Romano et al., 2003), have a higher proliferative capacity (Cotsarelis et al., 1989; Ebato et al.; Kruse et al.; Lindberg et al.; Pellegrini et al., 1999), and are slow-cycling as demonstrated by their ability to retain DNA labels such as tritiated thymidine (3H-TdR) over long periods of time (Cotsarelis et al., 1989). Under a steady state, the majority of LEPCs are “mitotically” quiescent and resting in the G0 phase, with only 4–5% of the LEPC population actively undergoing mitosis (Sagga et al., 2018),(Cotsarelis, Cheng et al. 1989). However, upon wounding of the corneal epithelium, LEPCs are activated to divide asymmetrically to produce two daughter cells: a stem cell, which remains in the limbal niche to replenish the stem cell pool for “self-renewal”, and a transient amplifying cell (TAC) that moves suprabasally in the limbus and centripetally into the cornea for “corneal fate decision” (Tseng, 1989); (Cotsarelis et al., 1989),(Di Girolamo et al., 2015). After a limited number of cell divisions, TAC differentiates into non-proliferative post-mitotic cells (PMCs), which lose contact with the basement membrane and migrate suprabasally as “terminally differentiated cells” (TDCs) (Pellegrini et al., 1999; Tseng, 1989) (Fig. 1D).
While a marker specific to LEPCs has yet to be confirmed, LEPCs can be distinguished from their progeny by a panel of markers including transporters such as ABCB5 and ATP binding cassette (ABCG2) (Ksander et al., 2014),(Watanabe et al., 2004); transcription factors such as Sox9 (Bath et al., 2013), C/EBPδ (Barbaro et al., 2007), Bmi-1 (Umemoto et al., 2006), Pax6 (Ramaesh and Dhillon, 2003), and p63α and its ΔNp63α isoform (Di Iorio et al., 2005; Shortt et al., 2007), cell adhesion molecules and receptors including Frizzled 7 (Mei et al., 2014), N-cadherin (Hayashi et al., 2007), and Notch-1 (Yoshida et al., 2006); and cytokeratins (CKs) such as CK15 (Yoshida et al., 2006), CK14 (Dua et al., 2005);(Ksander et al., 2014), CK3 (Schermer et al., 1986), and CK12 (Liu et al., 1994). Fig. 1 illustrates how these putative markers have been used to denote quiescence, self-renewal, and fate decision from LEPCs to TAC, PMC and TDC (Fig. 1E).
2. Neurovascular Support of Limbal Niche
Among all stem cell niches in the body, nerve fibers frequently coincide and coexist with blood vessels to form a neurovascular network. Taking the epidermis as an example, the upper bulge of the hair follicle, which harbors epidermal stem cells, is extensively innervated by cutaneous fibers (Larson et al., 2010). Whole mount immunostaining of the skin shows perfect alignment of blood vessels and nerves within a neurovascular niche (Larrivee et al., 2009). Sonic Hedgehog signaling pathway that mediates the sensory nerve influence on the epidermal stem cell niche in the upper bulge.(Larson et al., 2010) The unique expression of Gli1 by epidermal stem cells at the upper bulge depends on sensory nerves derived from dorsal root ganglion neurons expressing Sonic Hedgehog to wrap around it.(Larson et al., 2010) Surgical denervation selectively deprives the Gli1 expression resulting in a reduced number of bulge stem cells. A similar pattern of nerve-derived Sonic Hedgehog signaling is observed in controlling the taste bud density on the tongue,(Castillo et al., 2014) and renewal of touch dome stem cells in a perineural microenvironment in the epidermis. (Xiao et al., 2015)
Palisades of Vogt within the cornea limbus are extensively innervated (Lavker et al., 2004) and vascularized (Muller et al., 2003), suggesting that the neurovascular support is critical in maintaining limbal niche integrity. Sensory denervation of the ophthalmic branch of the trigeminal nerve leads to a 2 to 4-fold loss of limbal stem cells (Ueno et al., 2012). Similarly, capsaicin denervation impairs stem cell progeny egress from the hair follicle, resulting in delayed healing of denervated skin (Martinez-Martinez et al., 2012);(Gho et al., 2016). It is no wonder why insults to the limbal neurovascular network can lead to chronic inflammation as well as corneal pathologies including epithelial keratitis, epithelial defect, and stromal ulceration. For example, disruption of the first branch of the trigeminal nerve can cause neurotrophic keratitis, which overlaps with severe dry eye disease due to diminished blinking and tearing reflexes (Mead et al., 2020). Furthermore, the severity of chemical burns is graded by the degree of limbal ischemia (Dua et al., 2001). Nerve growth factor with its receptors TrkA and p75NTR are constitutively expressed in basal limbus and cornea. (Touhami et al., 2002),(Lambiase et al., 1998) and had shown to promote limbal stem cells phenotype and its proliferative capacity and clonal forming efficiency.(Kolli et al., 2019)Additional studies are needed to understand how the limbal niche is supported by its neurovascular network.
3. Limbal Niche Cells
In addition to its neurovascular network, the native limbal niche comprises extracellular matrix components and a subset of neighboring cells including stem cell progeny and subjacent mesenchymal cells (Fuchs et al., 2004). For the extracellular matrix, it has been shown that Laminin N-211, −213, −221, Vitronectin, Versican, BM40/SPARC, and Tenascin-C are more specific to the limbus than the cornea (Echevarria et al., 2011),(Schlotzer-Schrehardt et al., 2007);(Mei et al., 2012). Furthermore, a specialized hyaluronan (HA) matrix that differs from the rest of the cornea has been found in the murine limbal niche, and disruption of this matrix within the niche compromises corneal epithelial regeneration (Gesteira et al., 2017). The mesenchymal cells in the limbal niche include intraepithelial melanocytes, subepithelial mesenchymal cells, and migrating immune cells. Melanocytes, which are relatively concentrated in the limbal epithelial crypts, have been shown to maintain the undifferentiated state of epithelial progenitors during in vitro co-culture (Dziasko et al., 2015).
Our cumulative studies over the last decade have allowed us to conclude the following three salient features of a subset of mesenchymal cells that lie closely subjacent to limbal basal epithelial cells termed limbal niche cells (LNCs) in the limbal stroma (Table 1A):
Table 1.
Salient Features and Phenotypic Characterization of Limbal Niche Cells
| A. Salient Features | |
| |
| B. Phenotypic Characterization | |
| Embryonic Stem Cell Markers | Oct4, Sox2, Nanog, Rex1, SSEA4, CD34, ABCG2 (Chen et al 2011) |
| Angiogenesis Progenitor Markers | PDGFR-β, CD31, α-SMA, FLK-1 (Li et al 2012a) |
| Neural Crest Progenitor Markers | Pax6, Sox2, Nestin (Chen et al 2019),(Chen et al 2011) |
| Mesenchymal Stem Cells Markers | CD73, CD90, CD105 (Li et al 2012) |
The first salient feature is the physical close contact between LEPCs and LNCs in the vicinity of the limbal basement membrane. This feature explains why (1) the conventional isolation method utilizing dispase, which digests the basement membrane, has failed to isolate LNCs in the past, and (2) isolation the entire limbal stroma via trypsin may have included other mesenchymal cells deep into the stroma. In contrast, the isolation method based on collagenase, which digests interstitial collagens, but not the basement membrane, retains the close physical contact between LEPCs and LNCs so as to enable successful isolation of LNCs lying in the limbal epithelial crypt deep into the stroma (Chen et al., 2011). Such collagenase-isolated clusters obtained directly from an intact limbal tissue contain both Pancytokeratin (PCK) + / Vimentin - LEPCs that exhibit high clonogenicity and a subset of PCK − / Vimentin + mesenchymal cells termed LNCs (Chen et al., 2011). When freshly isolated, these LNCs are characterized as small as 10 μm in diameter and heterogeneously express embryonic stem cell (ESC) markers such as Oct4, Sox2, Nanog, Rex1, Nestin, N-cadherin, SSEA4 and CD34 (Chen et al., 2011), angiogenesis progenitor (pericyte) markers such as Platelet-derived growth factor receptor beta (PDGFR-β), VEGF receptor 2 (FLK-1), CD31, von Willebrand Factor (vWF), and α-SMA (Li et al., 2012a),(Li et al., 2012b), and neural crest progenitor markers such as Pax6, Sox2, Nestin, p75NTR, and Musashi-1 (Chen et al., 2019). Similar collagenase isolation method has been adopted by others to confirm the expression of ESC markers and derivation of neural crest progenitors after expansion (Basu et al., 2014; Chen et al., 2014b; Tomasello et al., 2016). Although the expressions of these and other markers has also been reported by collagenase digestion of the limbal stroma following manual scraping or dispase digestion of the overlying epithelial cells (Branch et al., 2012; Bray et al., 2014; Katikireddy et al., 2014), such methods may have unintentionally left some progenitor cells in the fraction retained by the filter due to tight association with the basement membrane matrix (Chen et al., 2014a). Table 1A summarizes markers identified for freshly isolated LNCs by us upon isolation and compared to those by others based on expansion after isolation using different enzymatic digestion methods from the human limbal stroma. LNC may also express neural cell crest markers such as HNK1, SOX9 and SOX10 when expanded in subsequent cultures.(Ghoubay-Benallaoua et al., 2017) Further studies are needed to determine the relationship between LNCs subjacent to the LEPCs and those lying deep in the stroma.
The second salient feature is the stringent ex vivo environment to maintain the in vivo LNC phenotype. We achieve this goal by culturing freshly-isolated LNCs on coated two-dimensional (2D) Matrigel in serum-free modified embryonic stem cell medium (MESCM) containing basic fibroblast growth factors (bFGF) and leukemia inhibitory factor (LIF) for up to 10 passages (Li et al., 2012a),(Li et al., 2012b). A number of studies have shown that corneal epithelial cells grown on relatively soft substrates are able to retain limbal stem cell markers, whereas cells cultured on stiff substrates are prone to differentiate (Foster et al., 2014; Gouveia et al., 2019; Jones et al., 2012; Moers et al., 2013). Others have also noted that increased matrix rigidity threatens mesenchymal stem cells and epithelial stem cell lineages (Abdeen et al., 2014; Engler et al., 2006; Wang and Chen, 2013). Although the expression of ESC markers and angiogenic progenitor markers by LNCs is gradually lost during serial passage on 2D Matrigel, such a loss is transient and can be regained when reseeded back on three-dimensional (3D) Matrigel (Li et al., 2012a; Xie et al., 2012). Besides matrix rigidity, we also noted that the aforementioned plasticity, i.e., reversibility by changing from 2D to 3D Matrigel, is irreversibly lost if LNCs are expanded in serum-containing medium that is traditionally used for mesenchymal stem cells expansion (Li et al., 2012b).
The third salient feature is that LNCs with an in vivo phenotype of nuclear Pax6+ neural crest progenitors have multipotent potential to differentiate into the classical tri-lineages of bone, cartilage and adipose tissue similar to mesenchymal stem cells (Li et al., 2012b), pericytes and angiogenic progenitors to promote angiogenesis (vasculogenesis),(Li et al., 2012a), and neurons, oligodendrocytes, and astrocytes (Chen et al., 2019) (Table 2). Others have also discovered similar multipotent potential in mesenchymal cells isolated from the human limbal stroma (Table 2). We discovered that nuclear Pax6+ staining is uniquely found in freshly-isolated LNCs but not corneal stromal cells (Chen et al., 2019). The hallmark of nuclear Pax6+ staining to denote the neural crest progenitor phenotype is causally correlated with neural crest marker expression, neutrosphere formation, and neuroglial differentiation (Chen et al., 2019). This finding is consistent with the notion that the limbal stroma cells are derived from migrating Pax-expressing neural crest during embryonic development (Baulmann et al., 2002). In fact, human cornea and limbal stromal cells have been reported to express neural crest genes such as Pax6 (Basu et al., 2014), ABCG2 (Du et al., 2005), Nestin (Chen et al., 2011), and Sox2 (Chen et al., 2011). Sox2-expressing neural crest progenitor cells have been found in the dermal papilla to support dermal follicular niche (Driskell et al., 2009). It is tempting to speculate that the multipotent potential of LNCs summarized in Table 2 supports their regenerative potential to restore the neurovascular network during wound healing. This speculation is supported by the finding that nestin-driven GFP cells that co-express with CD31 and vWF can grow into vessels from the bulge region of hair follicles during wounds healing (Amoh et al., 2004) and can subsequently be isolated and differentiated into neurons, glia, keratinocytes, smooth muscle, and melanocytes in vitro (Amoh et al., 2004). Although corneal epithelial differentiation by that limbal stromal neural crest progenitors has been suggested (Dravida et al., 2005),(Katikireddy et al., 2014), it has been challenged by the recent data from Ghoubay-Benallaoua et al 2017. (Ghoubay-Benallaoua et al., 2017)
Table 2.
Multipotent Potential of Mesenchymal Cells from Human Limbal Stroma
| MULTIPOTENT POTENTIAL | |
|---|---|
| Angiogenic Potential | References |
| Pericyte | (Li et al., 2012a)* |
| Vascular Endothelial Tube Formation | (Li et al., 2012a)* |
| Neuroglial Potential | |
| Neuron | (Dravida et al., 2005) (Chen et al., 2019)* |
| Astrocytes | (Chen et al., 2019)* |
| Oligodendrocytes | (Dravida et al., 2005)* |
| Neurospheres | (Ushida et al 2004) (Chen et al., 2019)*(Chen et al 2014) |
| Trilineage Potential | |
| Adipocytes | (Li et al 2012 ) *(Branch et al 2012) (Dravida et al., 2005) (Liang et al 2014) (Tomasello et al., 2016) |
| Osteocytes | (Li et al 2012)* (Dravida et al., 2005) (Liang et al 2014) (Tomasello et al., 2016) |
| Chondrocytes | (Li et al 2012)* (Dravida et al., 2005) (Liang et al 2014) (Tomasello et al., 2016) |
| Other Lineage Potentials | |
| Corneal Epithelia | (Dravida et al., 2005) (Gallagher et al., 2014) |
| Keratocytes | (Du et al., 2007) (Branch et al., 2012) (Basu et al., 2014) |
| Cardiac Myocytes | (Dravida et al., 2005) |
| Hepatocytes | (Dravida et al., 2005) |
| Pancreatic Islet cells | (Dravida et al., 2005) |
Published by Our Group
4. Regulatory Mechanism of LEPCs by LNCs
The in vivo close physical contact between LNCs and LEPCs can be recapitulated by an in vitro reunion assay by seeding single isolated LNC and LEPCs, which contain LEPC, at a 1:1 ratio on 3D Matrigel (Li et al., 2012a). A number of studies have shown that chemokine stromal cell-derived factor-1 (SDF-1, also known as CXCL12) is critical for the recruitment and retention of CXCR4+ bone marrow stem/progenitor cells to the neo-angiogenic niches to promote revascularization during wound healing (Petit et al., 2007; Petit et al., 2002; Ratajczak et al., 2004). We discovered that in vitro reunion of LNCs and LEPCs is also mediated by this chemokine axis (Xie et al., 2011). Specifically, we found that the SDF-1/CXCR4 axis is predominantly expressed in the human limbus but not the cornea and that SDF-1 is preferentially expressed by limbal epithelial cells and CXCR4 is expressed by subjacent stromal mesenchymal cells (Xie et al., 2011). Such reunion prevents fate decision of LEPCs, i.e., differentiation into corneal epithelial cells, upregulates p63α expression, and maintains holoclone growth signifying self-renewal of LEPCs (Fig. 2A). Disruption of such reunion by AMD3100, a small molecular inhibitor or a blocking antibody to CXCR4, at the time of seeding of both LNCs and LEPCs, but not later, promotes corneal fate decision and reduces holoclone growth of LEPCs (Xie et al., 2011). Furthermore, reunion between LEPCs with cells other than LNCs, such as remaining human limbal stromal cells, human mesenchymal stem cells, human corneal stromal cells, or human umbilical endothelial cells, leads to adoption of corneal epithelial fate (Xie et al., 2012);(Li et al., 2012a) (Fig. 2A). These findings collectively provide molecular mechanism thereby LNCs forms a close physical contact with LEPCs to maintain stem cell self-renewal and prevent corneal epithelial fate decision (Xie et al., 2011, 2012).
Figure 2. In Vitro Reunion of LNCs and LEPCs.

Reunion between LNCs and LEPCs in 3D Matrigel prevents corneal fate decision as illustrated by expression of cytokeratin 12 (CK12) by LEPCs as the corneal fate marker (A). Expression of CK12 is abolished when LNCs cultured in MESCM (Modified embryonic stem cells medium) are compared to reunion with RSC (remaining stromal cells after LNCs have been removed), bone marrow mesenchymal stem cells, human corneal fibroblast (HCF) or human umbilical endothelial cells (HUVEC) cultured in Endothelial Growth Media (EGM2), or LNC cultured in DF (DMEM/F12 with 10% FBS). In contrast, the expression of p63α as a marker for LEPCs is promoted (A). (Data taken from Li et al 2012a, Li et al 2012b) The reunion in 3D Matrigel promotes stem cell self-renewal by activating Wnt signaling but suppressing BMP signaling. In contrast, the reunion in HC-HA/PTX3 promotes stem cell quiescence by suppressing Wnt signaling but activating BMP signaling (B) LNC: Limbal Niche Cells; SC: Stem Cells (Quiescence or Self-Renewal); TAC: Transient Amplified Cells; TDC: Terminal Differentiated Cells. (Data taken from (Han et al 2014; Chen et al 2015)
Self-renewal and quiescence of stem cells are regulated by two segregated niche compartments through Wnt signaling pathways and bone morphogenetic protein (BMP) signaling pathways, respectively (Greco and Guo, 2010). We thus deployed the aforementioned in vitro reunion assay to see if these two signaling pathways also operate between LNCs and LEPCs. In 3D Matrigel, we noted marked clonal growth of LEPCs, signifying stem cell activation is correlated with activation of canonical Wnt signaling (Han et al., 2014). The involvement of Wnt signaling has been noted in the corneal fate decision controlled by Pax6 in human corneal epithelium where β-catenin is found in the perinuclear cytoplasm of LEPCs and correlated with upregulation of Wnt7A and FZD5 (Ouyang et al., 2014). Inactivation of BMP signaling in LNCs by noggin leads to downregulation of nuclear pSmad1/5/8 in LEPCs and nuclear β-catenin in larger LEPCs but membrane relocation of β-catenin in smaller LEPCs and significant upregulation of DKK1/2 (Han et al., 2014), suggesting the upregulation of DKK1 and DKK2 might suppress Wnt signaling in LEPCs adjacent to LNCs. These results collectively support the notion that the in vitro reunion of LNCs and LEPCs in 3D Matrigel recapitulates an in vitro limbal niche suggestive of stem cell self-renewal (Fig. 2B).
5. HC-HA/PTX3 from Amniotic Membrane as a Surrogate Matrix Niche
Since the reintroduction of amniotic membrane (AM) transplantation (AMT) in ophthalmology by Kim and Tseng in 1995 (Kim and Tseng, 1995), there has been a surge of interest in using this surgical procedure for ocular surface reconstruction (Fernandes et al., 2005; Liu et al., 2010; Sangwan et al., 2007; Sangwan and Tseng, 2001; Tseng, 2001), presumably due to the advent of cryopreservation, which devitalizes living cells to minimize immune reaction while preserving the remaining matrix integrity for long-term storage (Kruse et al., 2000). In 2001, the U.S. Food and Drug Administration (FDA) designated cryopreserved AM (Amniograft®; Bio-Tissue, Inc., Miami, FL) as a “361 human cell and tissue-based product (HCT/P)” for ocular wound repair and healing through exertion of anti-scarring and anti-inflammatory actions. Clinical use of cryopreserved AM is now considered the standard of care for many ophthalmic indications, is reimbursed by Centers for Medicare and Medicaid Services (CMS), and has over a 20-year safety record. Not only AM is useful for ocular surface reconstruction but also an efficient substrate for growing LSC with no 3T3 feeders as demonstrated by cultured cell transplantation (Sangwan et al., 2011),(Tsai et al., 2000),(Pauklin et al., 2010),(Borderie et al., 2019). Human AM epithelial cells can serve as feeder layers for promoting ex-vivo expansion of limbal epithelial progenitor cell.(Chen et al., 2007)
Our cumulative research effort over a period of more than a decade under the research support from the National Institute of Health has identified and characterized one key matrix component responsible for the aforementioned therapeutic actions of AM. We first demonstrated that the biological activity of cryopreserved AM is retained in a water-soluble AM extract. More specifically, we noted that this AM extract induces an anti-inflammatory effect by promoting apoptosis of activated macrophages (He et al., 2008; Li et al., 2006) and upregulating anti-inflammatory cytokines (i.e. IL-10) while downregulating pro-inflammatory markers (i.e. TNF-α, IL-6, CD86) (He et al., 2008). AM extract also exhibits an anti-scarring action by preventing myofibroblast expression of α-smooth muscle actin (a-SMA) (Li et al., 2008). We then utilized successive ultracentrifugation in a CsCl gradient in the presence of 4M guanidine HCl to isolate a unique HA-containing matrix from AM extract, termed HC-HA/PTX3, which consists of high molecular weight HA covalently linked with heavy chain 1 (HC1) from inter-α-trypsin inhibitor (IaI) through the catalytic action of TNFα-stimulated gene 6 (TSG-6) and is further complexed with pentraxin 3 (PTX3) (He et al., 2009; Zhang et al.) (Fig. 3). The biosynthetic process of HC-HA/PTX3 involves the following two steps: the first is to from HCHA complex via tumor necrosis factor-stimulated gene-6 (TSG-6), which is an enzyme that catalyzes the covalent (ester bond) transfer of HCs from IaI to HA.(Rugg et al., 2005);(Fulop et al., 2003);(Jessen and Odum, 2003);(Sanggaard et al., 2006) IαI contains two HCs (i.e., HC1 and HC2) and a light chain termed bikunin jointed a chondroitin sulfate chain and is present in the blood after being secreted by the liver.(Enghild et al., 1989);(Odum, 1990); (Salier et al., 1996);(Mizushima et al., 1998);(Blom et al., 1999);(Zhuo et al., 2004) We demonstrated that the HC-HA/PTX3 complex purified from AM consists of HMW HA (>3000 kDa) covalently linked with HC1 and tightly bound PTX3, but not HC2, bikunin, and TSG-6. Unlike the cumulus-oocyte complex, the source of IaI is endogenously produced by AM epithelial cells and stromal cells but not derived from the liver, and the expression of TSG-6 and PTX3 is constitutive (i.e., without relying on proinflammatory cytokines).(Blom et al., 1999),(Zhang et al., 2014) Similar to ovulation (Salustri et al., 2004);the second step is to from the HC-HA/PTX3 complex by tight association of the HC-HA complex with PTX3. Recent studies further disclose that HC-HA/PTX3 is a unique matrix component abundantly present in the birth tissue, i.e., not only in AM but also in umbilical cord (Tan et al., 2014),(Cooke et al., 2014).
Figure 3. Biosynthesis of HC-HA/PTX3.

HC-HA/PTX3 is synthesized by the following two steps: (1) In the first step, TNF-stimulated gene 6 protein (TSG-6) covalently binds heavy chain 1 (HC1) of inter-α-trypsin inhibitor (IαI) and transfers it to high molecular weight hyaluronan (HA), at which time HC1 becomes conjugated and TSG-6 released. (2) In the second step, PTX3, an octamer, is tightly associated with the HC-HA complex via binding with HCs. (Image Taken from (Tighe et al 2020) with modification.)
5.1. HC-HA/PTX3 Exerts Anti-Inflammatory and Anti-Scarring Effects
HC-HA/PTX3 exerts potent anti-inflammatory and anti-scarring actions. Similar to AM extract, HC-HA/PTX3 dose-dependently promotes apoptosis of activated but not resting macrophages (He et al., 2009; He et al., 2013) and suppresses activation of Th1 and Th17 lymphocytes (He et al., 2009), thus capable of modulating inflammation triggered by both innate and adaptive immune responses. Consequently, subconjunctival injection of HC-HA/PTX3 dose-dependently prolongs corneal allograft survival (He et al., 2014) and suppresses inflammation and scarring of both conjunctiva and lacrimal tissue thus preserving lacrimal tear production and conjunctival goblet cells in a murine model of graft-versus-host disease following allogeneic bone marrow transplantation (Ogawa, 2017). HC-HA/PTX3 also suppresses the TGF-β1 promoter activity of human corneal fibroblasts (He et al., 2009) and reverts human corneal fibroblasts and myofibroblasts to keratocytes by inhibiting canonical TGF-β signaling and activating BMP signaling (Zhu et al., 2020). Moreover, both AM extract and HC-HA/PTX3 suppress viability and tube formation of cultured HUVEC (Shay et al., 2011) but promote growth when HUVEC are cocultured with LNCs (unpublished observation), collectively suggesting that HC-HA/PTX3 suppresses abnormal angiogenesis mediated by endothelial cells alone but promotes vasculogenesis when endothelial cells are associated with pericytes.
5.2. HC-HA/PTX3 Preserves Quiescence of LEPCs through LNCs
The majority of LEPCs are “mitotically” quiescent and resting in the G0 phase under a steady state (Cotsarelis et al., 1989; Sagga et al., 2018). The G0 phase of quiescence stem cells has been assigned as a strategy for stem cells in rapid response to injury-induced signals (Rodgers et al 2014). Due to the complexity of factors in AM, purified HC-HA/PTX3 matrix helps delineate the potential mechanism explaining how AM might modulate interactions between LNCs and LEPCs.(Chen et al., 2015) Therefore, HC-HA/PTX3 may someday be deployed as a surrogate matrix niche in lieu of AM to support ex vivo expansion of LEPCs. Taking advantage of the aforementioned in vitro reunion assay, we have investigated whether stem cell quiescence can be maintained by HC-HA/PTX3 to replace 3D Matrigel. Others have reported that LEPCs use integrins α3β1 and α6β4 to anchor to their basement membrane niche (Polisetti et al., 2016), presumably for self-renewal. Our results showed that HC-HA/PTX3 promotes expression of ESC markers by LNCs more so than 3D Matrigel (Chen et al., 2015). Reunion of LNCs and LEPCs on HC-HA/PTX3 results in sphere growth that exhibits similar suppression of fate decision, i.e., preventing from differentiation into corneal epithelial cells but negligible clonal growth of LEPCs, which exhibit upregulation of quiescence markers including nuclear translocation of phosphorylated Bmi-1 (Chen et al., 2015). Furthermore, the above finding is correlated with the suppression of canonical Wnt but activation of noncanonical Wnt (PCP) signaling and BMP signaling in both LEPCs and LNCs. The activation of BMP signaling in LNCs is crucial because ablation of BMP signaling results in upregulation of cell cycle genes, downregulation of Bmi-1, nuclear exclusion of phosphorylated Bmi-1, and marked promotion of LEPC clonal growth (Chen et al., 2015). Hence, HC-HA/PTX3 uniquely preserves quiescence of LEPCs through the upregulation of BMP and non-canonical Wnt signaling in LNCs and subsequent BMP signaling in LEPCs (Fig. 2B). Therefore, the limbal niche resembles other stem cell niches such as dermal papillae, epidermal bulges, intestinal crypts, and bone marrow, where both Wnt and BMP signaling have been found to play an imperative role in two adjacent compartments that regulate stem cell self-renewal and quiescence, respectively (reviewed in (Li and Xie, 2005),(Greco and Guo, 2010; Li and Clevers, 2010). Along with the gradual loss of the expression of ESC markers and angiogenic progenitor markers (Li et al., 2012a; Xie et al., 2012), the nuclear Pax6+ neural crest progenitor status also gradually declines in LNCs following serial passage (Chen et al., 2019). Such a gradual loss can be reverted by seeding the late-passaged LNCs back to HC-HA/PTX3, but not 3D Matrigel, suggesting that HC-HA/PTX3 is capable of rejuvenating differentiated (aged) LNCs to restore their in vivo phenotype to support quiescence and self-renewal of LEPCs (Chen et al, 2020, manuscript submitted). Our recent data showed that activation of BMP signaling in LNCs proceeds after cell aggregation of LNCs mediated by SDF-1/CXCR4 signaling (Chen et al 2020 manuscript submitted), a finding that is also noted in human corneal fibroblasts when seeded on HC-HA/PTX3(Zhu et al., 2020). We speculate that this signaling is triggered by the binding between HC-HA/PTX3 and CD44, a well-known receptor for HA. If proven, it suggests that limbal stem cell niche uses different matrix ligand and receptor to control stem cell quiescence and self-renewal, respectively.
6. Clinical Evidence Supporting Amniotic Membrane as Surrogate Niche
Therefore, HC-HA/PTX3 as a single matrix can orchestrate a myriad of actions collectively translated to promote regenerative healing (reviewed in (Tseng, 2016; Tseng et al., 2016)). This notion has been demonstrated in the clinical efficacy of cryopreserved AM in promoting corneal nerve regeneration in patients suffering from moderate to severe dry eye (John et al., 2017) and chronic neuropathic pain (Morkin and Hamrah, 2018). We thus envisage that HC-HA/PTX3 is a unique matrix that can serve as a surrogate limbal niche. This notion is also supported by the finding that disruption of a specific HA matrix within the limbal niche leads to compromised corneal epithelial regeneration and more inflammatory response in knockout mice of HA synthetases or TSG-6 (Gesteira et al., 2017), which is involved in biosynthesis of HC-HA/PTX3 (Fig. 3). If LEPCs can be analogized as “seeds” to planted in the garden, the regenerative properties of HC-HA/PTX3 in supporting LNCs can be viewed as a key nutrient “fertilizer” that enriches the “top soil” (limbal niche) given the close interaction between LNCs and LEPCs (Fig. 4). This new paradigm is supported by the following clinical evidence:
Figure 4. HC-HA/PTX3 as a Surrogate Matrix Niche.

HC-HA/PTX3 exerts anti-inflammatory and anti-scarring actions, of which both act as “weeding” measures to fend off two untoward threats, i.e., chronic inflammation and scarring. HC-HA/PTX3 also acts as a “fertilizer” that provides essential nutrients to developing “seeds” (stem cells) and improves the quality of the “top soil” (limbal niche) through its supporting action to LNCs to maintain SC quiescence. Collectively, this myriad of actions collectively promote regenerative healing.
6.1. The Role of Pax6 in Pathogenesis of LSCD
Limbal stem cell deficiency (LSCD) is the pathological process leading to corneal blindness where the corneal tissues manifest chronic inflammation, scarring, vascularization, and conjunctivalization (see reviews in (Ahmad, 2012; Fernandez-Buenaga et al., 2018; Kim and Mian, 2017)). Besides direct destructive insults to reduce the LEPC pool by chemical/thermal burns, Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN), or contact lens wear, LSCD can also ensue from a dysfunctional limbal stromal niche. To understand a variety of etiologies of LSCD, readers might like to see the reviews and references cited therein (Hatch and Dana, 2009; Rossen et al., 2016; Vazirani et al., 2018);(Deng et al., 2019) Among diverse causes that may lead to a dysfunctional stromal niche, one prototypic disease is aniridia caused by haploinsufficiency mutations in the Pax6 gene (Cvekl and Callaerts, 2017). For the corneal epithelium, Pax6 plays an important role in maintaining epithelial proliferation (Collinson et al., 2004);(Hsueh et al., 2013), wound healing (Doran, 2008), and corneal fate decision (Ouyang et al., 2014). Nonetheless, Pax6 also plays a key role in maintaining the in vivo Pax6+ neural crest progenitor phenotype in LNCs that is crucial for supporting self-renewal and preventing fate decision (i.e., terminal differentiation) of LEPCs (Chen et al., 2019). Thus, the finding that HC-HA/PTX3 can help rejuvenate “aged” LNCs to regain the nuclear Pax6+ neural crest progenitor status may one day shed insight not only into the pathophysiology of LSCD caused by aniridia but also into the therapeutic potential of using HC-HA/PTX3-containing birth tissue (AM or umbilical cord) to restore and strengthen the supporting role of LNCs.
6.2. Amniotic Membrane Transplantation for Preventing and Treating LSCD
The idea that HC-HA/PTX3-containing birth tissue (AM or umbilical cord) can be used to restore and strengthen the supporting role of LNCs was first demonstrated when AM transplantation (AMT) was re-introduced in ophthalmology in 1995 (Kim and Tseng, 1995) in a rabbit model of LSCD induced by chemical and surgical insults. To a great surprise, that study disclosed that AMT prevented LSCD by restoring the normal corneal epithelial phenotype. Subsequently, this clinical effectiveness was demonstrated in human patients with “partial” LSCD, i.e., the cornea without total loss of LEPCs (Tseng et al., 1998),(Anderson et al., 2001);(Gomes et al., 2003);(Ivekovic et al., 2005); (Kheirkhah et al., 2008a);(Sharma et al., 2018)). Because AMT can prevent LSCD, especially if implemented within two weeks of acute chemical or thermal burns (Kheirkhah et al., 2008c; Meller et al., 2000; Prabhasawat et al., 2007), self-retained cryopreserved AM (e.g., PROKERA®; BioTissue, Inc., Miami, Florida, USA) was developed in 2004 to facilitate early intervention because its placement on the eye surface can readily be executed in-office or bed side. This sutureless approach prevents LSCD and subsequent cicatricial complications in managing acute chemical or thermal burns (Chirapapaisan et al., 2018; Kheirkhah et al., 2008c; Shupe and Cheng, 2017) as well as acute SJS/TENS (Kolomeyer et al., 2013; Shammas et al., 2010; Tomlins et al., 2013).
For treating one eye with the total loss of LEPCs in the setting of unilateral “total” LSCD, Kenyon and Tseng (Kenyon, 1989) first reported transplantation of 240° of conjunctival-limbal tissue from the healthy contralateral eye in a surgical procedure termed conjunctival limbal autograft (CLAU). Because of the concern that the donor eye might develop LSCD in the long run based on the experimental model (Chen and Tseng, 1990, 1991), others began to reduce the size of the harvested CLAU graft and noted that the clinical benefit in treating unilateral total LSCD vanishes when the size of CLAU is reduced to less than 90° (Moldovan et al., 1999; Rao et al., 1999). Because the experimental evidence shows that chronic inflammation within the limbal niche contributes to the CLAU failure (Tsai and Tseng, 1995), we and others have incorporated AMT with CLAU by taking the advantage of AM’s anti-inflammatory and anti-scarring actions (Ivekovic et al., 2005; Kheirkhah et al., 2008b; Meallet et al., 2003; Santos et al., 2005). Indeed, the addition of AMT has successfully reduced the size of CLAU to 60° for unilateral total LSCD (Kheirkhah et al., 2008d). Sangwan et al. (Sangwan et al., 2012) introduced simple limbal epithelial transplantation (SLET) where a small limbal graft is harvested from the healthy contralateral eye, cut into small pieces, and fixed with fibrin glue on an AM graft that covers the cornea for unilateral partial or total LSCD. The combined use of single (Sangwan et al., 2012) or double (Amescua et al., 2014) layers of AM in SLET has successfully reduced the size of limbal stem cell removal from the fellow eye down to less than 30° as testified by an overall success rate of 80% in a total of 378 cases during a long-term follow up (Jackson et al., 2020). Therefore, the new paradigm that AM can be used as a surrogate niche is also supported by the above clinical evidence showing successful in vivo expansion of LEPCs.
6.3. Ex Vivo Expansion (Tissue Engineering) of LEPCs
When treating patients with bilateral total LSCD, one would have to resort to transplantation of allogeneic LEPCs in a procedure termed keratolimbal allograft (Kim and Tseng, 1995). However, one key hurdle impeding the success of this procedure is allograft rejection, which is combated with long-term use of systemic immunosuppressive therapies (Liang et al., 2009). One way to circumvent this difficulty is by transplanting LEPCs following ex vivo expansion, a concept first introduced by Pellegrini et al. in 1997 using a culturing technique based on the use of mitomycin C-treated murine 3T3 fibroblast feeder layers (Pellegrini et al., 1997). Although ex vivo expansion was suggested to potentially reduce the risk of allograft rejection as antigen-presenting cells do not survive during culture (Shortt et al., 2011), this notion has not been demonstrated because systemic immunosuppression is still required according to long-term follow-up studies of cultured allogeneic limbal stem cells transplantation. (Shortt et al., 2014),(Borderie et al., 2019),(Pauklin et al., 2010). Ex vivo expansion of LEPCs has since been explored by many as a new therapeutic approach to address LSCD. They vary in methods of epithelial isolation from limbal biopsy, the use and preparation of AM as the substrate, the expansion medium, the use of airlifting to promote epithelial stratification, and the use of additional feeder layers such as mitomycin C-arrested murine 3T3 fibroblasts, mesenchymal stem cells, or human stromal cells (reviewed in (Tseng et al., 2010)). Now that we understand the importance of LNCs in supporting LEPC function, it is important to re-examine these variables by asking the question whether a surrogate niche has been restored and maintained to preserve the function of LEPCs during ex vivo expansion. If the said paradigm that HC-HA/PTX3 serves as a surrogate matrix niche holds true, one may like to build additional process controls to ensure HC-HA/PTX3 is preserved and matrix rigidity not jeopardizing the phenotype and function of LNCs during preparation or further manipulation of AM as the substrate (Borrelli et al., 2013),(Le and Deng, 2019). Furthermore, one may also like to pay attention to the preservation of the in vivo phenotype of nuclear Pax6+ neural crest progenitors in LNCs that can be threatened by the use of serum-containing media and serial passages.
7. Conclusion
In summary, the new paradigm that HC-HA/PTX3 or HC-HA/PTX3-containing birth tissue can serve as a surrogate matrix niche surely explains why AM plays a pivotal role in managing LSCD during in vivo or ex vivo expansion of LEPCs. Taking the metaphor of planting seeds in the garden (Figure 4), the anti-inflammatory and anti-scarring actions exerted by HC-HA/PTX3 act as “weeding” measures to fend untoward external insults to the niche, i.e., “soil”. The supportive action of HC-HA/PTX3 to LNCs, which in turn maintain stem cell quiescence and self-renewal, can be viewed as “fertilizer” that enriches the niche to become “top soil”. In conjunction with its anti-inflammatory and anti-scarring actions, HC-HA/PTX3 can promote regenerative healing (Tseng, 2016; Tseng et al., 2016). That is why such action paradigm has been successfully applied beyond ophthalmology as evidenced by the use of cryopreserved umbilical cord graft in promoting healing in complex diabetic foot ulcers (Caputo et al., 2016; Ghoubay-Benallaoua et al., 2017; Marston, 2019), radionecrotic wounds (Fernandez, 2019), and severe spina bifida (Papanna et al., 2016), preventing cicatricial complications in orthopedic reconstruction (DeMill, 2014) and expediting functional recovery of continence following robot-assisted radical prostatectomy (Ahmed et al., 2019). We thus envisage that the new paradigm based on HC-HA/PTX3 as a novel regenerative matrix to serve as a surrogate niche can set a new standard for regenerative medicine in the future.
Supplementary Material
Highlights:
Limbal niche nourishes, protects, and regulates quiescence, self-renewal, and differentiation decision of limbal epithelial stem cells (LESCs)
HC-HA/PTX3 found in amniotic membrane and umbilical cord may serve as a surrogate matrix niche to support quiescence and self-renewal but prevent corneal fate decision of LESCs
This may explain why amniotic membrane plays a pivotal role in managing limbal stem cell deficiency during in vivo or ex vivo expansion of LESCs
HC-HA/PTX3 as a surrogate niche can set a new standard for regenerative medicine in ophthalmology and beyond
Funding:
The work was supported by a RO1 EY06819 grant (to SCGT) from the National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Financial Disclosure: All authors are employees of TissueTech. Dr. Tseng has obtained a patent for the method of preparation and clinical uses of the birth tissue, i.e., amniotic membrane and umbilical cord, and has licensed the rights to TissueTech, Inc, which procures and processes, and to Bio-Tissue, Inc, and Amniox Medical, Inc., of which both are subsidiaries of TissueTech, Inc, to distribute cryopreserved birth tissue for clinical and research uses.
References
- Abdeen AA, Weiss JB, Lee J, Kilian KA, 2014. Matrix composition and mechanics direct proangiogenic signaling from mesenchymal stem cells. Tissue Eng Part A 20, 2737–2745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahmad S, 2012. Concise review: limbal stem cell deficiency, dysfunction, and distress. Stem Cells Transl Med 1, 110–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahmed M, Esposito M, Lovallo G, 2019. A single-center, retrospective review of robot-assisted laparoscopic prostatectomy with and 1 without cryopreserved umbilical cord allograft in improving continence recovery. Journal of robotic surgery In-press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amescua G, Atallah M, Nikpoor N, Galor A, Perez V, 2014. Modified simple limbal epithelial transplantation using cryopreserved amniotic membrane for unilateral limbal stem cell deficiency. Am J Ophthalmol 158, 469–475. [DOI] [PubMed] [Google Scholar]
- Amoh Y, Li L, Yang M, Moossa AR, Katsuoka K, Penman S, Hoffman RM, 2004. Nascent blood vessels in the skin arise from nestin-expressing hair-follicle cells. Proc Natl Acad Sci U S A 101, 13291–13295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anderson DF, Ellies P, Pires RT, Tseng SC, 2001. Amniotic membrane transplantation for partial limbal stem cell deficiency. Br J Ophthalmol 85, 567–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ang LP, Tan DT, Beuerman RW, Lavker RM, 2004. Development of a conjunctival epithelial equivalent with improved proliferative properties using a multistep serum-free culture system. Invest Ophthalmol Vis.Sci 45, 1789–1795. [DOI] [PubMed] [Google Scholar]
- Barbaro V, Testa A, Di IE, Mavilio F, Pellegrini G, De LM, 2007. C/EBPdelta regulates cell cycle and self-renewal of human limbal stem cells. J.Cell Biol 177, 1037–1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Basu S, Hertsenberg AJ, Funderburgh ML, Burrow MK, Mann MM, Du Y, Lathrop KL, Syed-Picard FN, Adams SM, Birk DE, Funderburgh JL, 2014. Human limbal biopsy-derived stromal stem cells prevent corneal scarring. Sci Transl Med 6, 266ra172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bath C, Muttuvelu D, Emmersen J, Vorum H, Hjortdal J, Zachar V, 2013. Transcriptional dissection of human limbal niche compartments by massive parallel sequencing. PloS one 8, e64244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baulmann DC, Ohlmann A, Flugel-Koch C, Goswami S, Cvekl A, Tamm ER, 2002. Pax6 heterozygous eyes show defects in chamber angle differentiation that are associated with a wide spectrum of other anterior eye segment abnormalities. Mech Dev 118, 3–17. [DOI] [PubMed] [Google Scholar]
- Blom AM, Morgelin M, Oyen M, Jarvet J, Fries E, 1999. Structural characterization of inter-alpha-inhibitor. Evidence for an extended shape. J Biol Chem. 274, 298–304. [DOI] [PubMed] [Google Scholar]
- Borderie VM, Ghoubay D, Georgeon C, Borderie M, de Sousa C, Legendre A, Rouard H, 2019. Long-Term Results of Cultured Limbal Stem Cell Versus Limbal Tissue Transplantation in Stage III Limbal Deficiency. Stem Cells Transl Med 8, 1230–1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Borrelli M, Reichl S, Feng Y, Schargus M, Schrader S, Geerling G, 2013. In vitro characterization and ex vivo surgical evaluation of human hair keratin films in ocular surface reconstruction after sterilization processing. J Mater Sci Mater Med 24, 221–230. [DOI] [PubMed] [Google Scholar]
- Branch MJ, Hashmani K, Dhillon P, Jones DR, Dua HS, Hopkinson A, 2012. Mesenchymal stem cells in the human corneal limbal stroma. Investigative ophthalmology & visual science 53, 5109–5116. [DOI] [PubMed] [Google Scholar]
- Bray LJ, Heazlewood CF, Munster DJ, Hutmacher DW, Atkinson K, Harkin DG, 2014. Immunosuppressive properties of mesenchymal stromal cell cultures derived from the limbus of human and rabbit corneas. Cytotherapy 16, 64–73. [DOI] [PubMed] [Google Scholar]
- Caputo WJ, Vaquero C, Monterosa A, Monterosa P, Johnson E, Beggs D, Fahoury GJ, 2016. A retrospective study of cryopreserved umbilical cord as an adjunctive therapy to promote the healing of chronic, complex foot ulcers with underlying osteomyelitis. Wound Repair Regen 24, 885–893. [DOI] [PubMed] [Google Scholar]
- Castillo D, Seidel K, Salcedo E, Ahn C, de Sauvage FJ, Klein OD, Barlow LA, 2014. Induction of ectopic taste buds by SHH reveals the competency and plasticity of adult lingual epithelium. Development 141, 2993–3002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen JJ, Tseng SC, 1990. Corneal epithelial wound healing in partial limbal deficiency. Investigative ophthalmology & visual science 31, 1301–1314. [PubMed] [Google Scholar]
- Chen JJ, Tseng SC, 1991. Abnormal corneal epithelial wound healing in partial-thickness removal of limbal epithelium. Investigative ophthalmology & visual science 32, 2219–2233. [PubMed] [Google Scholar]
- Chen SY, Cheng AMS, Zhang Y, Zhu YT, He H, Mahabole M, Tseng SCG, 2019. Pax 6 Controls Neural Crest Potential of Limbal Niche Cells to Support Self-Renewal of Limbal Epithelial Stem Cells. Sci Rep 9, 9763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen SY, Han B, Zhu YT, Mahabole M, Huang J, Beebe DC, Tseng SC, 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. [DOI] [PubMed] [Google Scholar]
- Chen SY, Hayashida Y, Chen MY, Xie HT, Tseng SC, 2011. A new isolation method of human limbal progenitor cells by maintaining close association with their niche cells. Tissue Eng Part C Methods 17, 537–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen SY, Mahabole M, Horesh E, Wester S, Goldberg JL, Tseng SC, 2014a. Isolation and characterization of mesenchymal progenitor cells from human orbital adipose tissue. Invest Ophthalmol.Vis.Sci 55, 4842–4852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X, Thomson H, Cooke J, Scott J, Hossain P, Lotery A, 2014b. Adult limbal neurosphere cells: a potential autologous cell resource for retinal cell generation. PloS one 9, e108418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen YT, Li W, Hayashida Y, He H, Chen SY, Tseng DY, Kheirkhah A, Tseng SC, 2007. Human amniotic epithelial cells as novel feeder layers for promoting ex vivo expansion of limbal epithelial progenitor cells. Stem Cells 25, 1995–2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng AM, Chua L, Casas V, Tseng SC, 2016. Morselized Amniotic Membrane Tissue for Refractory Corneal Epithelial Defects in Cicatricial Ocular Surface Diseases. Translational vision science & technology 5, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chirapapaisan C, Prabhasawat P, Srivannaboon S, Roongpoovapatr V, Chitsuthipakorn P, 2018. Ocular Injury due to Potassium Permanganate Granules. Case Rep Ophthalmol 9, 132–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Collinson JM, Chanas SA, Hill RE, West JD, 2004. Corneal development, limbal stem cell function, and corneal epithelial cell migration in the Pax6(+/−) mouse. Investigative ophthalmology & visual science 45, 1101–1108. [DOI] [PubMed] [Google Scholar]
- Cooke M, Tan EK, Mandrycky C, He H, O’Connell J, Tseng SC, 2014. Comparison of cryopreserved amniotic membrane and umbilical cord tissue with dehydrated amniotic membrane/chorion tissue. J.Wound.Care 23, 465–476. [DOI] [PubMed] [Google Scholar]
- Cotsarelis G, Cheng SZ, Dong G, Sun T-T, Lavker RM, 1989. Existence of slow-cycling limbal epithelial basal cells that can be preferentially stimulated to proliferate: implications on epithelial stem cells. Cell 57, 201–209. [DOI] [PubMed] [Google Scholar]
- Cvekl A, Callaerts P, 2017. PAX6: 25th anniversary and more to learn. Experimental eye research 156, 10–21. [DOI] [PubMed] [Google Scholar]
- DeMill SL, Granata JD, McAlister JE, Berlet GC, Hyer CF, 2014. Safety analysis of cryopreserved amniotic membrane/umbilical cord tissue in foot and ankle surgery: a consecutive case series of 124 patients. Surgical Technology Internation 25, 257–261. [PubMed] [Google Scholar]
- Deng SX, Borderie V, Chan CC, Dana R, Figueiredo FC, Gomes JAP, Pellegrini G, Shimmura S, Kruse FE, and The International Limbal Stem Cell Deficiency Working, G., 2019. Global Consensus on Definition, Classification, Diagnosis, and Staging of Limbal Stem Cell Deficiency. Cornea 38, 364–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Girolamo N, Bobba S, Raviraj V, Delic NC, Slapetova I, Nicovich PR, Halliday GM, Wakefield D, Whan R, Lyons JG, 2015. Tracing the fate of limbal epithelial progenitor cells in the murine cornea. Stem Cells 33, 157–169. [DOI] [PubMed] [Google Scholar]
- Di Iorio E, Barbaro V, Ruzza A, Ponzin D, Pellegrini G, De Luca M, 2005. Isoforms of DeltaNp63 and the migration of ocular limbal cells in human corneal regeneration. Proc Natl Acad Sci U S A 102, 9523–9528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doran M, 2008. Amniotic Membrane for Acute Stevens-Johnson Syndrome, EyeNet. [Google Scholar]
- Dravida S, Pal R, Khanna A, Tipnis SP, Ravindran G, Khan F, 2005. The transdifferentiation potential of limbal fibroblast-like cells. Brain Res.Dev.Brain Res 160, 239–251. [DOI] [PubMed] [Google Scholar]
- Driskell RR, Giangreco A, Jensen KB, Mulder KW, Watt FM, 2009. Sox2-positive dermal papilla cells specify hair follicle type in mammalian epidermis. Development 136, 2815–2823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du Y, Funderburgh ML, Mann MM, SundarRaj N, Funderburgh JL, 2005. Multipotent stem cells in human corneal stroma. Stem Cells 23, 1266–1275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du Y, Sundarraj N, Funderburgh ML, Harvey SA, Birk DE, Funderburgh JL, 2007. Secretion and organization of a cornea-like tissue in vitro by stem cells from human corneal stroma. Investigative ophthalmology & visual science 48, 5038–5045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dua HS, King AJ, Joseph A, 2001. A new classification of ocular surface burns. Br J Ophthalmol 85, 1379–1383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dua HS, Shanmuganathan VA, Powell-Richards A, Tiqhe PJ, Joseph A, 2005. Limbal epithelial crypts: a novel anatomical structure and a putative limbal stem cell niche. Br J Ophthalmol 89, 529–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dziasko MA, Tuft SJ, Daniels JT, 2015. Limbal melanocytes support limbal epithelial stem cells in 2D and 3D microenvironments. Experimental eye research 138, 70–79. [DOI] [PubMed] [Google Scholar]
- Ebato B, Friend J, Thoft RA, 1987. Comparison of central and peripheral human corneal epithelium in tissue culture. Invest.Ophthalmol.Vis.Sci 28, 1450–1456. [PubMed] [Google Scholar]
- Echevarria TJ, Chow S, Watson S, Wakefield D, Di Girolamo N, 2011. Vitronectin: a matrix support factor for human limbal epithelial progenitor cells. Investigative ophthalmology & visual science 52, 8138–8147. [DOI] [PubMed] [Google Scholar]
- Enghild JJ, Thogersen IB, Pizzo SV, Salvesen G, 1989. Analysis of inter-alpha-trypsin inhibitor and a novel trypsin inhibitor, pre-alpha-trypsin inhibitor, from human plasma. Polypeptide chain stoichiometry and assembly by glycan. J Biol Chem. 264, 15975–15981. [PubMed] [Google Scholar]
- Engler AJ, Sen S, Sweeney HL, Discher DE, 2006. Matrix elasticity directs stem cell lineage specification. Cell 126, 677–689. [DOI] [PubMed] [Google Scholar]
- Fernandes M, Sridhar MS, Sangwan VS, Rao GN, 2005. Amniotic membrane transplantation for ocular surface reconstruction. Cornea 24, 643–653. [DOI] [PubMed] [Google Scholar]
- Fernandez-Buenaga R, Aiello F, Zaher SS, Grixti A, Ahmad S, 2018. Twenty years of limbal epithelial therapy: an update on managing limbal stem cell deficiency. BMJ open ophthalmology 3, e000164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernandez D, 2019. Cryopreserved amniotic membrane and umbilical cord for a radiation-induced wound with exposed dura: a case report. Journal of wound care 28, S4–s8. [DOI] [PubMed] [Google Scholar]
- Foster JW, Jones RR, Bippes CA, Gouveia RM, Connon CJ, 2014. Differential nuclear expression of Yap in basal epithelial cells across the cornea and substrates of differing stiffness. Experimental eye research 127, 37–41. [DOI] [PubMed] [Google Scholar]
- Fuchs E, Tumbar T, Guasch G, 2004. Socializing with the neighbors: stem cells and their niche. Cell 116, 769–778. [DOI] [PubMed] [Google Scholar]
- Fulop C, Szanto S, Mukhopadhyay D, Bardos T, Kamath RV, Rugg MS, Day AJ, Salustri A, Hascall VC, Glant TT, Mikecz K, 2003. Impaired cumulus mucification and female sterility in tumor necrosis factor-induced protein-6 deficient mice. Development 130, 2253–2261. [DOI] [PubMed] [Google Scholar]
- Gallagher C, Clarke C, Aherne ST, Katikireddy KR, Doolan P, Lynch V, Shaw S, Bobart-Hone A, Murphy C, Clynes M, Power W, O’Sullivan F, 2014. Comparative transcriptomic analysis of cultivated limbal epithelium and donor corneal tissue reveals altered wound healing gene expression. Investigative ophthalmology & visual science 55, 5795–5805. [DOI] [PubMed] [Google Scholar]
- Gesteira TF, Sun M, Coulson-Thomas YM, Yamaguchi Y, Yeh LK, Hascall V, Coulson-Thomas VJ, 2017. Hyaluronan Rich Microenvironment in the Limbal Stem Cell Niche Regulates Limbal Stem Cell Differentiation. Investigative ophthalmology & visual science 58, 4407–4421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gho CG, Schomann T, de Groot SC, Frijns JH, Rivolta MN, Neumann MH, Huisman MA, 2016. Isolation, expansion and neural differentiation of stem cells from human plucked hair: a further step towards autologous nerve recovery. Cytotechnology 68, 1849–1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghoubay-Benallaoua D, de Sousa C, Martos R, Latour G, Schanne-Klein MC, Dupin E, Borderie V, 2017. Easy xeno-free and feeder-free method for isolating and growing limbal stromal and epithelial stem cells of the human cornea. PloS one 12, e0188398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goldberg MF, Bron AJ, 1982. Limbal palisades of Vogt. Trans.Am.Ophthalmol.Soc 80, 155–171. [PMC free article] [PubMed] [Google Scholar]
- Gomes JA, dos Santos MS, Cunha MC, Mascaro VL, Barros JN, de Sousa LB, 2003. Amniotic membrane transplantation for partial and total limbal stem cell deficiency secondary to chemical burn. Ophthalmology 110, 466–473. [DOI] [PubMed] [Google Scholar]
- Gouveia RM, Lepert G, Gupta S, Mohan RR, Paterson C, Connon CJ, 2019. Assessment of corneal substrate biomechanics and its effect on epithelial stem cell maintenance and differentiation. Nat Commun 10, 1496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greco V, Guo S, 2010. Compartmentalized organization: a common and required feature of stem cell niches? Development 137, 1586–1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grieve K, Ghoubay D, Georgeon C, Thouvenin O, Bouheraoua N, Paques M, Borderie VM, 2015. Three-dimensional structure of the mammalian limbal stem cell niche. Experimental eye research 140, 75–84. [DOI] [PubMed] [Google Scholar]
- Han B, Chen SY, Zhu YT, Tseng SC, 2014. Integration of BMP/Wnt signaling to control clonal growth of limbal epithelial progenitor cells by niche cells. Stem Cell Res. 12, 562–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hatch KM, Dana R, 2009. The structure and function of the limbal stem cell and the disease states associated with limbal stem cell deficiency. International ophthalmology clinics 49, 43–52. [DOI] [PubMed] [Google Scholar]
- Hayashi R, Yamato M, Sugiyama H, Sumide T, Yang J, Okano T, Tano Y, Nishida K, 2007. N-Cadherin is expressed by putative stem/progenitor cells and melanocytes in the human limbal epithelial stem cell niche. Stem Cells 25, 289–296. [DOI] [PubMed] [Google Scholar]
- He H, Li W, Chen SY, Zhang S, Chen YT, Hayashida Y, Zhu YT, Tseng SC, 2008. Suppression of activation and induction of apoptosis in RAW264.7 cells by amniotic membrane extract. Investigative ophthalmology & visual science 49, 4468–4475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He H, Li W, Tseng DY, Zhang S, Chen SY, Day AJ, Tseng SC, 2009. Biochemical characterization and function of complexes formed by hyaluronan and the heavy chains of inter-alpha-inhibitor (HC*HA) purified from extracts of human amniotic membrane. J Biol Chem 284, 20136–20146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He H, Tan Y, Duffort S, Perez VL, Tseng SC, 2014. In vivo downregulation of innate and adaptive immune responses in corneal allograft rejection by HC-HA/PTX3 complex purified from amniotic membrane. Investigative ophthalmology & visual science 55, 1647–1656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He H, Zhang S, Tighe S, Son J, Tseng SC, 2013. Immobilized Heavy Chain-Hyaluronic Acid Polarizes Lipopolysaccharide-activated Macrophages toward M2 Phenotype. J Biol Chem. 288, 25792–25803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsueh YJ, Kuo PC, Chen JK, 2013. Transcriptional regulators of the DeltaNp63: their role in limbal epithelial cell proliferation. J Cell Physiol 228, 536–546. [DOI] [PubMed] [Google Scholar]
- Ivekovic R, Tedeschi-Reiner E, Novak-Laus K, ndrijevic-Derk B, Cima I, Mandic Z, 2005. Limbal graft and/or amniotic membrane transplantation in the treatment of ocular burns. Ophthalmologica 219, 297–302. [DOI] [PubMed] [Google Scholar]
- Jackson CJ, Myklebust Erno IT, Ringstad H, Tonseth KA, Dartt DA, Utheim TP, 2020. Simple limbal epithelial transplantation: Current status and future perspectives. Stem Cells Transl Med 9, 316–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jessen TE, Odum L, 2003. Role of tumour necrosis factor stimulated gene 6 (TSG-6) in the coupling of inter-alpha-trypsin inhibitor to hyaluronan in human follicular fluid. Reproduction. 125, 27–31. [DOI] [PubMed] [Google Scholar]
- John T, Tighe S, Sheha H, Hamrah P, Salem Z, Cheng A, Wang M, Rock N, 2017. Corneal Nerve Regeneration after Self-Retained Cryopreserved Amniotic Membrane in Dry Eye Disease. Journal of Ophthalmology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones RR, Hamley IW, Connon CJ, 2012. Ex vivo expansion of limbal stem cells is affected by substrate properties. Stem Cell Res 8, 403–409. [DOI] [PubMed] [Google Scholar]
- Katikireddy KR, Dana R, Jurkunas UV, 2014. Differentiation potential of limbal fibroblasts and bone marrow mesenchymal stem cells to corneal epithelial cells. Stem Cells 32, 717–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kenyon KR, 1989. Limbal autograft transplantation for chemical and thermal burns. Dev.Ophthalmol 18, 53–58. [DOI] [PubMed] [Google Scholar]
- Kheirkhah A, Casas V, Raju VK, Tseng SC, 2008a. Sutureless amniotic membrane transplantation for partial limbal stem cell deficiency. Am J Ophthalmol 145, 787–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kheirkhah A, Casas V, Sheha H, Raju VK, Tseng SC, 2008b. Role of conjunctival inflammation in surgical outcome after amniotic membrane transplantation with or without fibrin glue for pterygium. Cornea 27, 56–63. [DOI] [PubMed] [Google Scholar]
- Kheirkhah A, Johnson DA, Paranjpe DR, Raju VK, Casas V, Tseng SC, 2008c. Temporary sutureless amniotic membrane patch for acute alkaline burns. Arch Ophthalmol 126, 1059–1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kheirkhah A, Raju VK, Tseng SC, 2008d. Minimal conjunctival limbal autograft for total limbal stem cell deficiency. Cornea 27, 730–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim JC, Tseng SC, 1995. Transplantation of preserved human amniotic membrane for surface reconstruction in severely damaged rabbit corneas. Cornea 14, 473–484. [PubMed] [Google Scholar]
- Kim KH, Mian SI, 2017. Diagnosis of corneal limbal stem cell deficiency. Curr Opin Ophthalmol 28, 355–362. [DOI] [PubMed] [Google Scholar]
- Kolli S, Bojic S, Ghareeb AE, Kurzawa-Akanbi M, Figueiredo FC, Lako M, 2019. The Role of Nerve Growth Factor in Maintaining Proliferative Capacity, Colony-Forming Efficiency, and the Limbal Stem Cell Phenotype. Stem Cells 37, 139–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kolomeyer AM, Do BK, Tu Y, Chu DS, 2013. Placement of ProKera in the management of ocular manifestations of acute Stevens-Johnson syndrome in an outpatient. Eye & contact lens 39, e7–11. [DOI] [PubMed] [Google Scholar]
- Kruse FE, Joussen AM, Rohrschneider K, You L, Sinn B, Baumann J, Volcker HE, 2000. Cryopreserved human amniotic membrane for ocular surface reconstruction. Graefe’s Arch.Clin.Exp.Ophthalmol 238, 68–75. [DOI] [PubMed] [Google Scholar]
- Kruse FE, Kach JM, Klein U, Rohrschneider K, Vokker HE, Waubke TN, 1993. The importance of limbal stem cells for the therapy of chemical burns. Ger.J.Ophthalmol 2, 289. [Google Scholar]
- Ksander BR, Kolovou PE, Wilson BJ, Saab KR, Guo Q, Ma J, McGuire SP, Gregory MS, Vincent WJ, Perez VL, Cruz-Guilloty F, Kao WW, Call MK, Tucker BA, Zhan Q, Murphy GF, Lathrop KL, Alt C, Mortensen LJ, Lin CP, Zieske JD, Frank MH, Frank NY, 2014. ABCB5 is a limbal stem cell gene required for corneal development and repair. Nature 511, 353–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kulkarni BB, Tighe PJ, Mohammed I, Yeung AM, Powe DG, Hopkinson A, Shanmuganathan VA, Dua HS, 2010. Comparative transcriptional profiling of the limbal epithelial crypt demonstrates its putative stem cell niche characteristics. BMC.Genomics 11, 526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lambiase A, Bonini S, Micera A, Rama P, Bonini S, Aloe L, 1998. Expression of nerve growth factor receptors on the ocular surface in healthy subjects and during manifestation of inflammatory diseases. Invest.Ophthalmol.Vis.Sci 39, 1272–1275. [PubMed] [Google Scholar]
- Larrivee B, Freitas C, Suchting S, Brunet I, Eichmann A, 2009. Guidance of vascular development: lessons from the nervous system. Circ Res 104, 428–441. [DOI] [PubMed] [Google Scholar]
- Larson BJ, Longaker MT, Lorenz HP, 2010. Scarless fetal wound healing: a basic science review. Plast Reconstr Surg. 126, 1172–1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lavker RM, Tseng SC, Sun TT, 2004. Corneal epithelial stem cells at the limbus: looking at some old problems from a new angle. Experimental eye research 78, 433–446. [DOI] [PubMed] [Google Scholar]
- Le Q, Deng SX, 2019. The application of human amniotic membrane in the surgical management of limbal stem cell deficiency. The ocular surface 17, 221–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li GG, Chen SY, Xie HT, Zhu YT, Tseng SC, 2012a. Angiogenesis potential of human limbal stromal niche cells. Investigative ophthalmology & visual science 53, 3357–3367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li GG, Zhu YT, Xie HT, Chen SY, Tseng SC, 2012b. Mesenchymal stem cells derived from human limbal niche cells. Investigative ophthalmology & visual science 53, 5686–5697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li L, Clevers H, 2010. Coexistence of quiescent and active adult stem cells in mammals. Science 327, 542–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li L, Xie T, 2005. Stem cell niche: structure and function. Annu.Rev.Cell Dev.Biol 21, 605–631. [DOI] [PubMed] [Google Scholar]
- Li W, He H, Chen YT, Hayashida Y, Tseng SC, 2008. Reversal of myofibroblasts by amniotic membrane stromal extract. J Cell Physiol 215, 657–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W, He H, Kawakita T, Espana EM, Tseng SCG, 2006. Amniotic membrane induces apoptosis of interferon-gamma activited macrophages in vitro. Exp Eye Res. 82, 282–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang L, Sheha H, Tseng SC, 2009. Long-term outcomes of keratolimbal allograft for total limbal stem cell deficiency using combined immunosuppressive agents and correction of ocular surface deficits. Arch.Ophthalmol 127, 1428–1434. [DOI] [PubMed] [Google Scholar]
- Lindberg K, Brown ME, Chaves HV, Kenyon KR, Rheinwald JG, 1993. In vitro preparation of human ocular surface epithelial cells for transplantation. Invest.Ophthalmol.Vis.Sci 34, 2672–2679. [PubMed] [Google Scholar]
- Liu C-Y, Zhu G, Converse R, Kao CW-C, Nakamura H, Tseng SCG, Mui M-M, Seyer J, Justice MJ, Stech ME, Hansen GM, Kao WW-Y, 1994. Characterization and chromosomal localization of the cornea-specific murine keratin gene Krt1.12. J.Biol.Chem 269, 24627–24636. [PubMed] [Google Scholar]
- Liu J, Sheha H, Fu Y, Liang L, Tseng SC, 2010. Update on amniotic membrane transplantation. Expert.Rev.Ophthalmol 5, 645–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marston e.a., 2019. An open-label trial of cryopreserved human umbilical cord in the treatment of complex diabetic foot ulcers complicated by osteomyelitis. Wound Repair Regen. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinez-Martinez E, Galvan-Hernandez CI, Toscano-Marquez B, Gutierrez-Ospina G, 2012. Modulatory role of sensory innervation on hair follicle stem cell progeny during wound healing of the rat skin. PloS one 7, e36421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mead O, Tighe S, Tseng S, 2020. Amniotic Membrane Transplantation for Managing Dry Eye and Neurotrophic Keratitis. TJO. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meallet MA, Espana EM, Grueterich M, Ti SE, Goto E, Tseng SC, 2003. Amniotic membrane transplantation with conjunctival limbal autograft for total limbal stem cell deficiency. Ophthalmology 110, 1585–1592. [DOI] [PubMed] [Google Scholar]
- Mei H, Gonzalez S, Deng SX, 2012. Extracellular Matrix is an Important Component of Limbal Stem Cell Niche. J Funct Biomater 3, 879–894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mei H, Nakatsu MN, Baclagon ER, Deng SX, 2014. Frizzled 7 maintains the undifferentiated state of human limbal stem/progenitor cells. Stem Cells 32, 938–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meller D, Pires RTF, Mack RJS, Figueiredo F, Heiligenhaus A, Park WC, Prabhasawat P, John T, McLeod SD, Steuhl KP, Tseng SCG, 2000. Amniotic membrane transplantation for acute chemical or thermal burns. Ophthalmology 107, 980–990. [DOI] [PubMed] [Google Scholar]
- Mizushima S, Nii A, Kato K, Uemura A, 1998. Gene expression of the two heavy chains and one light chain forming the inter-alpha-trypsin-inhibitor in human tissues. Biol Pharm Bull 21, 167–169. [DOI] [PubMed] [Google Scholar]
- Moers K, Steinberg T, Schlunck G, Reinhard T, Tomakidi P, Eberwein P, 2013. Substrate elasticity as biomechanical modulator of tissue homeostatic parameters in corneal keratinocytes. Exp Cell Res 319, 1889–1901. [DOI] [PubMed] [Google Scholar]
- Moldovan SM, Borderie V, Baudrimont M, Laroche L, 1999. Treatment of unilateral limbal stem cell deficiency syndrome by limbal autograft. J.Fr.Ophtalmol 22, 302–309. [PubMed] [Google Scholar]
- Morkin MI, Hamrah P, 2018. Efficacy of self-retained cryopreserved amniotic membrane for treatment of neuropathic corneal pain. The ocular surface 16, 132–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muller LJ, Marfurt CF, Kruse F, Tervo TM, 2003. Corneal nerves: structure, contents and function. Exp.Eye Res 76, 521–542. [DOI] [PubMed] [Google Scholar]
- Niederer RL, Perumal D, Sherwin T, McGhee CN, 2007. Age-related differences in the normal human cornea: a laser scanning in vivo confocal microscopy study. Br J Ophthalmol 91, 1165–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Notara M, Shortt AJ, O’Callaghan AR, Daniels JT, 2013. The impact of age on the physical and cellular properties of the human limbal stem cell niche. Age (Dordr) 35, 289–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Odum L, 1990. Inter-alpha-trypsin inhibitor and pre-alpha-trypsin inhibitor in health and disease. Determination by immunoelectrophoresis and immunoblotting. Biol Chem Hoppe Seyler 371, 1153–1158. [PubMed] [Google Scholar]
- Ogawa R, 2017. Keloid and Hypertrophic Scars Are the Result of Chronic Inflammation in the Reticular Dermis. Int J Mol Sci 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ouyang H, Xue Y, Lin Y, Zhang X, Xi L, Patel S, Cai H, Luo J, Zhang M, Zhang M, Yang Y, Li G, Li H, Jiang W, Yeh E, Lin J, Pei M, Zhu J, Cao G, Zhang L, Yu B, Chen S, Fu XD, Liu Y, Zhang K, 2014. WNT7A and PAX6 define corneal epithelium homeostasis and pathogenesis. Nature 511, 358–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Papanna R, Fletcher S, Moise KJ Jr., Mann LK, Tseng SC, 2016. Cryopreserved Human Umbilical Cord for In Utero Myeloschisis Repair. Obstetrics and gynecology 128, 325–330. [DOI] [PubMed] [Google Scholar]
- Pauklin M, Fuchsluger TA, Westekemper H, Steuhl KP, Meller D, 2010. Midterm results of cultivated autologous and allogeneic limbal epithelial transplantation in limbal stem cell deficiency. Dev Ophthalmol 45, 57–70. [DOI] [PubMed] [Google Scholar]
- Pellegrini G, Golisano O, Paterna P, Lambiase A, Bonini S, Rama P, De Luca M, 1999. Location and clonal analysis of stem cells and their differentiated progeny in the human ocular surface. J.Cell Biol 145, 769–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pellegrini G, Traverso CE, Franzi AT, Zingirian M, Cancedda R, De Luca M, 1997. Long-term restoration of damaged corneal surface with autologous cultivated corneal epithelium. Lancet (London, England) 349, 990–993. [DOI] [PubMed] [Google Scholar]
- Petit I, Jin D, Rafii S, 2007. The SDF-1-CXCR4 signaling pathway: a molecular hub modulating neo-angiogenesis. Trends Immunol. 28, 299–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petit I, Szyper-Kravitz M, Nagler A, Lahav M, Peled A, Habler L, Ponomaryov T, Taichman RS, Arenzana-Seisdedos F, Fujii N, Sandbank J, Zipori D, Lapidot T, 2002. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol 3, 687–694. [DOI] [PubMed] [Google Scholar]
- Polisetti N, Zenkel M, Menzel-Severing J, Kruse FE, Schlotzer-Schrehardt U, 2016. Cell Adhesion Molecules and Stem Cell-Niche-Interactions in the Limbal Stem Cell Niche. Stem Cells 34, 203–219. [DOI] [PubMed] [Google Scholar]
- Prabhasawat P, Tesavibul N, Prakairungthong N, Booranapong W, 2007. Efficacy of amniotic membrane patching for acute chemical and thermal ocular burns. J Med.Assoc.Thai 90, 319–326. [PubMed] [Google Scholar]
- Ramaesh K, Dhillon B, 2003. Ex vivo expansion of corneal limbal epithelial/stem cells for corneal surface reconstruction. Eur.J Ophthalmol 13, 515–524. [DOI] [PubMed] [Google Scholar]
- Rao SK, Rajagopal R, Sitalakshmi G, Padmanabhan P, 1999. Limbal autografting: comparison of results in the acute and chronic phases of ocular surface burns. Cornea 18, 164–171. [DOI] [PubMed] [Google Scholar]
- Ratajczak MZ, Kucia M, Reca R, Majka M, Janowska-Wieczorek A, Ratajczak J, 2004. Stem cell plasticity revisited: CXCR4-positive cells expressing mRNA for early muscle, liver and neural cells ‘hide out’ in the bone marrow. Leukemia 18, 29–40. [DOI] [PubMed] [Google Scholar]
- Romano AC, Espana EM, Yoo SH, Budak MT, Wolosin JM, Tseng SC, 2003. Different cell sizes in human limbal and central corneal basal epithelia measured by confocal microscopy and flow cytometry. Invest Ophthalmol.Vis.Sci 44, 5125–5129. [DOI] [PubMed] [Google Scholar]
- Rossen J, Amram A, Milani B, Park D, Harthan J, Joslin C, McMahon T, Djalilian A, 2016. Contact Lens-induced Limbal Stem Cell Deficiency. The ocular surface 14, 419–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rugg MS, Willis AC, Mukhopadhyay D, Hascall VC, Fries E, Fulop C, Milner CM, Day AJ, 2005. Characterization of complexes formed between TSG-6 and inter-alpha-inhibitor that act as intermediates in the covalent transfer of heavy chains onto hyaluronan. J Biol Chem. 280, 25674–25686. [DOI] [PubMed] [Google Scholar]
- Sagga N, Kuffova L, Vargesson N, Erskine L, Collinson JM, 2018. Limbal epithelial stem cell activity and corneal epithelial cell cycle parameters in adult and aging mice. Stem Cell Res 33, 185–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salier J-P, Rouet P, Raguenez G, Daveau M, 1996. The inter-a-inhibitor family: from structure to regulation. Biochem J 315, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salustri A, Garlanda C, Hirsch E, De Acetis M, Maccagno A, Bottazzi B, Doni A, Bastone A, Mantovani G, Peccoz PB, 2004. PTX3 plays a key role in the organization of the cumulus oophorus extracellular matrix and in in vivo fertilization. Development 131, 1577–1586. [DOI] [PubMed] [Google Scholar]
- Sanggaard KW, Sonne-Schmidt CS, Jacobsen C, Thogersen IB, Valnickova Z, Wisniewski HG, Enghild JJ, 2006. Evidence for a two-step mechanism involved in the formation of covalent HC x TSG-6 complexes. Biochemistry 45, 7661–7668. [DOI] [PubMed] [Google Scholar]
- Sangwan VS, Basu S, MacNeil S, Balasubramanian D, 2012. Simple limbal epithelial transplantation (SLET): a novel surgical technique for the treatment of unilateral limbal stem cell deficiency. Br J Ophthalmol 96, 931–934. [DOI] [PubMed] [Google Scholar]
- Sangwan VS, Basu S, Vemuganti GK, Sejpal K, Subramaniam SV, Bandyopadhyay S, Krishnaiah S, Gaddipati S, Tiwari S, Balasubramanian D, 2011. Clinical outcomes of xeno-free autologous cultivated limbal epithelial transplantation: a 10-year study. Br J Ophthalmol 95, 1525–1529. [DOI] [PubMed] [Google Scholar]
- Sangwan VS, Burman S, Tejwani S, Mahesh SP, Murthy R, 2007. Amniotic membrane transplantation: a review of current indications in the management of ophthalmic disorders. Indian J.Ophthalmol 55, 251–260. [DOI] [PubMed] [Google Scholar]
- Sangwan VS, Tseng SC, 2001. New perspectives in ocular surface disorders. An integrated approach for diagnosis and management 1. Indian J.Ophthalmol 49, 153–168. [PubMed] [Google Scholar]
- Santos MS, Gomes JA, Hofling-Lima AL, Rizzo LV, Romano AC, Belfort R Jr., 2005. Survival analysis of conjunctival limbal grafts and amniotic membrane transplantation in eyes with total limbal stem cell deficiency. Am.J.Ophthalmol 140, 223–230. [DOI] [PubMed] [Google Scholar]
- Schermer A, Galvin S, Sun TT, 1986. Differentiation-related expression of a major 64K corneal keratin in vivo and in culture suggests limbal location of corneal epithelial stem cells. J Cell Biol 103, 49–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schlotzer-Schrehardt U, Dietrich T, Saito K, Sorokin L, Sasaki T, Paulsson M, Kruse FE, 2007. Characterization of extracellular matrix components in the limbal epithelial stem cell compartment. Exp.Eye Res 85, 845–860. [DOI] [PubMed] [Google Scholar]
- Shammas MC, Lai EC, Sarkar JS, Yang J, Starr CE, Sippel KC, 2010. Management of acute Stevens-Johnson syndrome and toxic epidermal necrolysis utilizing amniotic membrane and topical corticosteroids. Am J Ophthalmol 149, 203–213 e202. [DOI] [PubMed] [Google Scholar]
- Sharma N, Mohanty S, Jhanji V, Vajpayee RB, 2018. Amniotic membrane transplantation with or without autologous cultivated limbal stem cell transplantation for the management of partial limbal stem cell deficiency. Clinical ophthalmology (Auckland, N.Z.) 12, 2103–2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shay E, He H, Sakurai S, Tseng SC, 2011. Inhibition of angiogenesis by HC.HA, a complex of hyaluronan and the heavy chain of inter-alpha-inhibitor, purified from human amniotic membrane. Invest Ophthalmol.Vis.Sci 52, 2669–2678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shortt AJ, Bunce C, Levis HJ, Blows P, Dore CJ, Vernon A, Secker GA, Tuft SJ, Daniels JT, 2014. Three-year outcomes of cultured limbal epithelial allografts in aniridia and Stevens-Johnson syndrome evaluated using the Clinical Outcome Assessment in Surgical Trials assessment tool. Stem Cells Transl Med 3, 265–275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shortt AJ, Secker GA, Munro PM, Khaw PT, Tuft SJ, Daniels JT, 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, 1402–1409. [DOI] [PubMed] [Google Scholar]
- Shupe AT, Cheng AMS, 2017. Case report: Use of self-retained cryopreserved amniotic membrane in the treatment of acute chemical ocular burn. Optom Vis Sci 94, 1062–1065. [DOI] [PubMed] [Google Scholar]
- Tan EK, Cooke M, Mandrycky C, Mahabole M, He H, O’Connell J, McDevitt TC, Tseng SCG, 2014. Structural and Biological Comparison of Cryopreserved and Fresh Amniotic Membrane Tissues. Journal of Biomaterials and Tissue Engineering 4, 379–388. [Google Scholar]
- Tomasello L, Musso R, Cillino G, Pitrone M, Pizzolanti G, Coppola A, Arancio W, Di Cara G, Pucci-Minafra I, Cillino S, Giordano C, 2016. Donor age and long-term culture do not negatively influence the stem potential of limbal fibroblast-like stem cells. Stem Cell Res Ther 7, 83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tomlins PJ, Parulekar MV, Rauz S, 2013. “Triple-TEN” in the Treatment of Acute Ocular Complications From Toxic Epidermal Necrolysis. Cornea 32, 365–369. [DOI] [PubMed] [Google Scholar]
- Touhami A, Grueterich M, Tseng SC, 2002. The role of NGF signaling in human limbal epithelium expanded by amniotic membrane culture. Invest Ophthalmol.Vis.Sci 43, 987–994. [PubMed] [Google Scholar]
- Townsend WM, 1991. The limbal palisades of Vogt 1. Trans.Am.Ophthalmol.Soc 89, 721–756. [PMC free article] [PubMed] [Google Scholar]
- Tsai RJ, Li L, Chen J, 2000. Reconstruction of damaged corneas by transplantation of autologous limbal epithelial cells(1). Am.J.Ophthalmol 130, 543. [DOI] [PubMed] [Google Scholar]
- Tsai RJF, Tseng SCG, 1995. Effect of stromal inflammation on the outcome of limbal transplantation for corneal surface reconstruction. Cornea 14, 439–449. [PubMed] [Google Scholar]
- Tseng S, 2016. HC-HA/PTX3 Purified from Amniotic Membrane as Novel Regenerative Matrix: Insight into Relationship between Inflammation and Regeneration. IOVS 57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tseng SC, 2001. Amniotic membrane transplantation for ocular surface reconstruction. Biosci.Rep 21, 481–489. [DOI] [PubMed] [Google Scholar]
- Tseng SC, Chen SY, Shen YC, Chen WL, Hu FR, 2010. Critical appraisal of ex vivo expansion of human limbal epithelial stem cells. Curr Mol Med 10, 841–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tseng SC, He H, Zhang S, Chen SY, 2016. Niche Regulation of Limbal Epithelial Stem Cells: Relationship between Inflammation and Regeneration. The ocular surface 14, 100–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tseng SC, Prabhasawat P, Barton K, Gray T, Meller D, 1998. Amniotic membrane transplantation with or without limbal allografts for corneal surface reconstruction in patients with limbal stem cell deficiency. Arch Ophthalmol 116, 431–441. [DOI] [PubMed] [Google Scholar]
- Tseng SCG, 1989. Concept and application of limbal stem cells. Eye 3, 141–157. [DOI] [PubMed] [Google Scholar]
- Ueno H, Ferrari G, Hattori T, Saban DR, Katikireddy KR, Chauhan SK, Dana R, 2012. Dependence of corneal stem/progenitor cells on ocular surface innervation. Investigative ophthalmology & visual science 53, 867–872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Umemoto T, Yamato M, Nishida K, Yang J, Tano Y, Okano T, 2006. Limbal epithelial side-population cells have stem cell-like properties, including quiescent state. Stem Cells 24, 86–94. [DOI] [PubMed] [Google Scholar]
- Vazirani J, Nair D, Shanbhag S, Wurity S, Ranjan A, Sangwan V, 2018. Limbal Stem Cell Deficiency-Demography and Underlying Causes. Am J Ophthalmol 188, 99–103. [DOI] [PubMed] [Google Scholar]
- Wang YK, Chen CS, 2013. Cell adhesion and mechanical stimulation in the regulation of mesenchymal stem cell differentiation. J Cell Mol Med 17, 823–832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watanabe A, Yokoi N, Kinoshita S, Hino Y, Tsuchihashi Y, 2004. Clinicopathologic study of conjunctivochalasis. Cornea 23, 294–298. [DOI] [PubMed] [Google Scholar]
- Wong DWK, Chan W-K, Tan DTH, 1997. Harvesting a lamellar graft from a corneoscleral button: A new technique. Am.J.Ophthalmol, 688–689. [DOI] [PubMed] [Google Scholar]
- Xiao Y, Thoresen DT, Williams JS, Wang C, Perna J, Petrova R, Brownell I, 2015. Neural Hedgehog signaling maintains stem cell renewal in the sensory touch dome epithelium. Proc Natl Acad Sci U S A 112, 7195–7200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie HT, Chen SY, Li GG, Tseng SC, 2011. Limbal epithelial stem/progenitor cells attract stromal niche cells by SDF-1/CXCR4 signaling to prevent differentiation. Stem Cells 29, 1874–1885. [DOI] [PubMed] [Google Scholar]
- Xie HT, Chen SY, Li GG, Tseng SC, 2012. Isolation and expansion of human limbal stromal niche cells. Investigative ophthalmology & visual science 53, 279–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang PT, Sharpen L, Chan A, Chan CC, 2014. Indications for amniotic membrane grafts used in Ontario from 2011 to 2012. Can J Ophthalmol 49, 399–399. [DOI] [PubMed] [Google Scholar]
- Yoshida S, Shimmura S, Kawakita T, Miyashita H, Den S, Shimazaki J, Tsubota K, 2006. Cytokeratin 15 can be used to identify the limbal phenotype in normal and diseased ocular surfaces. Invest Ophthalmol Vis.Sci 47, 4780–4786. [DOI] [PubMed] [Google Scholar]
- Zhang S, He H, Day AJ, Tseng SC, 2012. Constitutive expression of inter-alpha-inhibitor (IalphaI) family proteins and tumor necrosis factor-stimulated gene-6 (TSG-6) by human amniotic membrane epithelial and stromal cells supporting formation of the heavy chain-hyaluronan (HC-HA) complex. J Biol Chem. 287, 12433–12444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang S, Zhu YT, Chen SY, He H, Tseng SC, 2014. Constitutive expression of pentraxin 3 (PTX3) protein by human amniotic membrane cells leads to formation of the heavy chain (HC)-hyaluronan (HA)-PTX3 complex. J Biol Chem. 289, 13531–13542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng TY, Xu JJ, 2008. [Evolving concepts on limbal stem cells and their niche]. Zhonghua Yan Ke Za Zhi 44, 469–472. [PubMed] [Google Scholar]
- Zhu YT, Li F, Zhang Y, Chen SY, Tighe S, Lin SY, Tseng SCG, 2020. HC-HA/PTX3 Purified From Human Amniotic Membrane Reverts Human Corneal Fibroblasts and Myofibroblasts to Keratocytes by Activating BMP Signaling. Investigative ophthalmology & visual science 61, 62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhuo L, Hascall VC, Kimata K, 2004. Inter-alpha-trypsin inhibitor, a covalent protein-glycosaminoglycan-protein complex. J Biol Chem. 279, 38079–38082. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
