Role of Stem Cells in Lacrimal and Meibomian Gland ...

4 downloads 0 Views 1MB Size Report
members of the microRNA-200 family (miR-200a, miR-. 200b, miR-200c, miR-141 and miR-429) and miR-205 were markedly downregulated in epithelial cells ...
CHAPTER

Driss Zoukhri Helen P. Makarenkova

13

Role of Stem Cells in Lacrimal and Meibomian Gland Development and Regeneration

INTRODUCTION The non-keratinized epithelia of the ocular surface are constantly challenged by environmental insults such as smoke, dust, or airborne pathogens. Protection of eye surface is mediated, in part by the secretions of two major glands, the lacrimal (LG) and meibomian (MG). In humans, the LG is the primary contributor to the aqueous layer of the tear film. The meibomian gland (MG) is responsible for the production of meibum, an oily secretion that retards evaporation of the preocular tear film. When LG and MG secretions decrease, or the composition of their secretions change, it affects the health of the ocular surface. The LG and MG are dynamic tissues that are capable of responding to physiological stimuli (i.e., environmental insults or severe injury) by mounting a well-orchestrated regenerative response that restores their cytoarchitecture. However, the mechanisms of LG and MG regeneration and the source of progenitor cells are still not very well defined. Here, we provide a timely review of LG and MG development, maintenance, and repair with the focus on emerging areas in stem cell research.

DISORDERS OF THE TEAR FILM DUE TO LACRIMAL AND MEIBOMIAN GLAND DISEASES Tears are the sole physical protective barrier for the ocular surface. The preocular tear film can be divided into three interacting layers: 1) an inner mucous layer that coats the cornea and conjunctiva; 2) an aqueous middle layer; and 3) an outer lipid layer which floats on the aqueous layer. Each layer of the tear film is secreted by a different set of glands.1-3 The mucous layer is secreted by the conjunctival goblet cells,4 the lipid layer is secreted by the MG that line the eyelid,1 and the aqueous layer is secreted by the main and accessory LGs.2 Production of tears in insufficient quantity or of inadequate quality results in constant irritation of the ocular surface leading to dry eye disease also referred to as keratoconjunctivitis sicca (KCS).5 Dry eye disease secondary to LG deficiency is known as aqueousdeficient type of dry eye.5 The second major class of dry eye is called evaporative dry eye and is mainly due to MG deficiency.5,6

1

STEM CELLS IN OPHTHALMOLOGY Patients with KCS can experience intense pain due to eye irritation, gritty/scratchy feeling in the eyes, blurry vision and light sensitivity.5 LG dysfunction that occurs as a result of autoimmunity, such as Sjögren’s syndrome or rheumatoid arthritis, accounts for an estimated 1-4 million patients in the United States suffering from dry eye syndromes.7-9 Furthermore, KCS occurring in the absence of an underlying autoimmune disease is most prevalent in people over the age of 50 and postmenopausal women.10,11 Furthermore, dry eye due to MG dysfunction is thought to account for a large number of the dry eye patient population. Recent epidemiological studies reported that an estimated 25-30 million Americans suffer from dry eye syndromes.5,10-15 These numbers are expected to rise significantly in the next decades with the aging population.5,13 Aqueous, as well as evaporative dry eye, are usually linked with inflammation-induced damage to the lacrimal and meibomian glands with consequent immune-mediated destruction of the glandular secretory units. Therefore, the question arises as to how one could halt or better yet reverse this cellular loss and trigger tissue regeneration. Recent studies conducted using various animal models suggest that both glands are capable of repair following experimentally induced injury and that they contain stem/progenitor cells that are recruited during the repair phase.

STRUCTURE AND DEVELOPMENT OF THE LG The LG develops through branching morphogenesis. In branching morphogenesis, repetitive epithelial cleft and bud formation creates complex three-dimensional branching structures. The LG has both epithelial and mesenchymal components16,17 and its morphogenesis involves a series of reciprocal interactions between epithelium and mesenchyme that result in the formation of a three-dimensional tubular network.18 The epithelial component of the LG is of tubulo-acinar morphology and consists of three cell lineages: ductal, acinar and myoepithelial cells. Acinar cells of the LG secrete mucous, seromucous, and serous.19 LG mesenchyme is derived from periorbital cells of neural crest origin. Several growth factors, including fibroblast growth factors 7 and 10 (FGF7, FGF10) and bone morphogenetic protein 7 (BMP7) have been shown to be the main regulators of LG branching

2

morphogenesis.16-18,20-22 Thus, over-expression of FGF7 or 10 in the embryonic cornea induces differentiation of corneal epithelial cells into functional lacrimal gland epithelium.20,21 These studies suggest that correct spatial and temporal expression of FGFs plays a critical role in normal lacrimal gland induction and development.23,24 Human LG consists of two lobes, which are both located superior to the orbit on the temporal side. One lobe is orbital, while the other lies in the palpebral region. There are multiple lacrimal duct openings in the dorsal eyelid.25 The development of the human LG has been studied extensively.26-28 LG morphogenesis in humans starts at O’Rahilly’s stages 19–20, by a thickening of the superior fornix epithelium near mesenchymal condensation. At the O’Rahilly’s stage 21, the first epithelial bud invaginates into the surrounding mesenchyme and later at stage 23, lumina appear within the glandular epithelial buds.27 The gland maturates during the weeks 9–16, acquiring morphology of adult LG.27 In mice, the two lobes of the LG are located differently. The mature LG also consists of a small intraorbital lobe and a much larger exorbital lobe situated adjacent to the external auditory apparatus. Both lobes are connected by a single duct that opens onto the rim of the lower eyelid at the temporal terminus.17 Using a transgene reporter construct based on an ectodermal enhancer from the Pax6 gene (P6 5.0-lacZ) we were able to characterize murine LG development in great detail16,17,20,29-31 (Figure 1). The first morphological indication of murine LG development is an ectodermal invagination from the conjunctival epithelium at the temporal edge of the eye (Figure 1A-D) while nasal invagination of the conjunctival epithelium (Figure 1E) gives rise to harderian gland. The lacrimal bud extends away from the eye but remains as a single bud until E15.5 (Figure 1F). The first branches in the developing gland appear between E15.5 and E16.5 (Figure 1G) and by E19.5, further branches are apparent in both the intraand exorbital lobes.17

LG REGENERATION AND EPITHELIAL MESENCHYMAL TRANSITION LG has a high regenerative potential and is able to repair itself even after substantial damage.32 LG regeneration is accompanied by a process called epithelial mesenchymal transition (EMT). During EMT, epithelial cells lose cell-cell attachment, polarity and epithelial-specific markers, undergo cytoskeletal

CHAPTER 13: Role of Stem Cells in Lacrimal and Meibomian Gland Development and Regeneration

Figure 1 - Lacrimal gland development. (A) Section of the eye in an E12.5 P6 5.0 lacZ reporter mouse. lacZ expression is indicated by the blue X-gal labeling in the lens epithelium (le) and the future conjunctival epithelium on the temporal side of the eye (red arrowhead). (B) Head region of E13.5 P6 5.0 lacZ reporter mouse showing X-gal-labeled corneal epithelium and lens. The lacrimal bud (red arrowhead) is observed projecting towards the auditory canal (ac). (C) As in B, but at higher magnification. The X-gal labeled lens epithelium (l) can be seen through the cornea, the epithelium (c) of which is also labeled. The lacrimal gland bud (red arrowhead) projects form the X-gal-labeled conjunctival epithelium. (D and E) Histological sections through the eye in an E13.5 P6 5.0 lacZ reporter mouse. (D) Blue X-gal labeling is apparent in the continuous layer of the embryonic corneal and conjunctival epithelium (ce) and the early LG bud (red arrowhead) is seen extending from the deepest aspect of the conjunctival epithelium (the fornix). (E) Reporter construct expression is absent from the fornix on the nasal side (black arrowhead). Early budding of the harderian gland primordium is apparent on the surface of the epithelium on the retinal side and is also negative for reporter expression (blue arrowhead). (F) The tip of the lacrimal bud has extended to a position dorsal to the stapedial artery (sa) and caudal to the bifurcation of its supraorbital (dashed lines – sas) and infraorbital (dashed lines – sai) branches by E15.5. (G) By E16.5 the first branching events are apparent. Adapted from Makarenkova et al., 2000.

remodeling, and gain a mesenchymal phenotype.33-36 Several reports suggest that epithelial cells of tissues undergoing EMT might have a certain level of plasticity and are able to dedifferentiate and then redifferentiate when regeneration is completed.37,38 However, it still remains unclear how much stem/progenitor cells or EMT contributes to the regeneration process.

tissue remodeling during embryogenesis39 (Figure 2). The role of EMT in tissue repair/regeneration is well described. Several studies reported the occurrence of EMT during tissue repair in several tissues including mammary glands, liver, kidney, and lung.40-46 It is generally well accepted that induction of EMT generates cells with mesenchymal stem-like properties.41,47,48

Epithelial-Mesenchymal Transition During LG Regeneration

Down-regulation of E-cadherin gene expression, an adherens junction protein, is critical for initiation of EMT and is associated with loss of cell polarity.33,34,36 Several transcription factors have been shown to regulate EMT.35,36,39 Thus, Snai1 seems to be a master regulator

Epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) play major roles in

3

STEM CELLS IN OPHTHALMOLOGY of EMT and acts partly by repressing expression of epithelial markers, such as E-cadherin and induction of mesenchymal markers, such as vimentin expression.46,49-51

and transcription factors controlling the expression of MMPs at different stages of LG development or disease progression.

It was recently shown that EMT is activated during repair of murine LG and generates cells with mesenchymal phenotype referred to as mesenchymal stem cells (MSC) that migrate to the site of injury and initiate LG repair.52 It was suggested that, during the later phases of LG repair, the MET process is initiated and MSC differentiate and form acinar and ductal epithelial cells.52 MSCs can be isolated from injured LGs and propagated in vitro. However, additional studies are needed to characterize the molecular mechanisms controlling both EMT and MET during LG repair. Such studies could unravel novel strategies to restore functional LG tissue and hence adequate production of the aqueous layer of the tear film.

We recently reported that during LG development mesenchymally expressed FGF10 and epithelially expressed homeobox transcription factor Barx2 cooperate in regulation of epithelially and mesenchymally expressed MMPs. Moreover, we, and others found that Barx2 binds to regulatory sequences of different MMPs and controls epithelial cell migration through the ECM.53 Our findings that BMPs regulate Barx2 expression54 and that BMPs, FGFs, and Barx2 regulate MMP expression and control Notch signaling55-57 and cell proliferation, migration and differentiation in different cellular contexts suggest that this signaling pathway may be involved in EMT and could be considered for pharmacological intervention.

Although EMT is essential for embryonic development and regeneration of many exocrine glands it can also be destructive for the epithelium and could possibly lead to dry eye conditions or tumors if the mechanism or timing of EMT is deregulated. It is also known that LG of Sjögren’s syndrome patients do not regenerate efficiently, suggesting that this disease prevents reverse transition of the MSC, i.e., MET. Thus, defining the molecular mechanisms controlling EMT and MET processes would be extremely useful for development of new strategies to treat some types of dry eye conditions.

Several recent studies suggest that specific MicroRNAs (miRNAs) are involved in regulation of the EMT/MET58 in different tissues. MiRNAs are small non-coding RNAs that target multiple messenger RNAs (mRNAs) and function as post-transcriptional regulators of gene expression regulating many biological processes including embryogenesis, regeneration, and EMT. Recently, the miRNA-dependent post-transcriptional regulation of development-associated genes was proposed to play a role in regulation of tissue regeneration and EMT initiation. Several up-to-date potential microRNA modulators of EMT have been identified. Thus, all five members of the microRNA-200 family (miR-200a, miR200b, miR-200c, miR-141 and miR-429) and miR-205 were markedly downregulated in epithelial cells that had undergone EMT.59 It was reported that dysregulation of miR-204 and/or miR200 mediates EMT and migration and invasion of endometrial and lung cancer.58,60 MiR-204 is an important endogenous negative regulator of Runx2 and can act as a regulator of EMT during secondary cataract formation.61 Moreover, TGF-β receptor 2 (TGF-β R2) and Snail2 (found to be important for induction of MET in LG) are direct targets of miR204.62 These findings suggest that downregulation of microRNAs may be an important step in EMT-mediated epithelial tissue regeneration.

Role of Mesenchymal Cells in Controlling the LG EMT/MET Epithelial structure of the LG is maintained by cellcell and cell extracellular matrix (ECM) interactions. Mesenchymal cells contribute to formation and maintenance of the ECM by production of different matrix components or matrix remodeling enzymes such as MMPs. Mesenchymal cells are also a source to multiple signaling molecules that can bind the ECM and control and guide epithelial growth. For example, several FGFs, and specifically FGF10, regulate all stages of LG morphogenesis. These findings suggest that signals leading to EMT within the LG could be produced by mesenchymal cells. The necessity to control the proteolytic activity of MMPs for treatment of diseases such as cancer or Sjögren’s syndrome has been acknowledged, however little progress has been made in this direction. The reason for this could be due to our limited knowledge of signaling pathways

4

LG REGENERATION AND STEM/ PROGENITOR CELLS

CHAPTER 13: Role of Stem Cells in Lacrimal and Meibomian Gland Development and Regeneration Evidence for the Presence of LG Stem Cells The LG is composed of multiple cell types including epithelial, mesenchymal, neuronal, and endothelial cell lineages that are essential for normal physiological function and maintenance of the gland. At the same time, secretory function of the LG completely depends on development and differentiation of LG epithelial components. Lacrimal and salivary glands are ectodermal appendages that develop through similar cellular mechanisms that involve an intimate crosstalk between the epithelium and mesenchyme. However, mutations that perturb primary epithelial bud formation leads to the complete absence of LG epithelial component and therefore secretory cells, while the mesenchymal component of the gland is still present.17,24,63,64 These findings suggest that the epithelial component of the LG is essential for development of a functional LG. In addition, this suggests that epithelial and mesenchymal components of the LG might contain separate lineage specific stem/progenitor cells. Both LG and salivary glands are organs with a high risk of being damaged due to their constant secretory activity, and it becomes extremely important that they retain their ability to regenerate during the whole life span. Based on their similar epithelial origin, LG and submanidibular glands (SMG) have many common characteristics, and as suggested in recent publications, may also have a similar putative stem cell population that gives rise to multipotent stem/progenitor cells in each epithelial organ.65 Several studies indicate that multipotent progenitor cells are present in the lacrimal gland and are involved in their regeneration.66-68 However, the nature of these cells is still not well defined, with more recent studies indicating that myoepithelial cells (MECs) and/or MCSs may have the capacity to act as stem cells and mediate regeneration.66-68 In addition to MECs and ductal epithelial cells, progenitor populations have been proposed but not identified.69,70 Stem cells are generally considered to be slowly dividing cells and are the precursors of progenitor cells, which are more proliferative, lineage-committed, with less capacity to self-renew, and may be lineage-specific. The common method for identifying slow cycling cells consists of using the 5-bromo-2’-deoxyuridine (BrdU) pulse-chase technique. BrdU is a thymidine analog that incorporates into the DNA of dividing cells (during the S phase of the cell cycle), rendering them detectable by immunohistochemical means. Subsequent cell

divisions in the absence of label (chase period) dilute the incorporated BrdU and only cells with the lowest replication profile could be detected.71-73 In a recent study, we showed that LG contains the BrdU-label retaining slow cycling cells that are mobilized to proliferate during LG repair.66 These findings support the idea that slow cycling stem cells with regeneration potential are present within the adult LG. LG Epithelial Progenitor Cells Acinar cell: LG acinar cells are highly polarized cells that secrete proteins, electrolytes and water. Although LG acinar cells are fully differentiated highly specialized secretory cells, they have a certain level of plasticity. Thus, highly purified preparations of murine lacrimal acinar cells cultured in Matrigel rafts resulted in the formation of functional acini that mimic function of acinar cells in uninjured LG.74-76 Another group was able to induce the efficient proliferation of primary rabbit lacrimal gland acinar cells by treating cells with epidermal growth factor (EGF) and dihydrotestosterone (DHT).75 A recent study of human LGs also suggests the presence of stem cells within the acinar cells.77 Although it is possible that LG acinar cells have their own stem/ progenitor cells, this hypothesis needs to be supported by lineage tracing experiments. Thus, isolated LG acinar cells may still contain centroacinar cells. In fact centroacinar cells (found within majority of endocrine glands) are an extension of the intercalated duct cells closely associated with the gland acini and were shown to possess stem cell properties.78-81 Myoepithelial cells: (MECs) are also found in multiple glandular organs such as LG, salivary, harderian, sweat and mammary glands. They surround glandular secretory epithelium, express SMA, and can contract helping secretory function of these exocrine glands.82-87 In the LG and other exocrine glands, MECs synthesize the basement membrane and form a functional network around the acinar and ductal cells separating them from the basement membrane and the mesenchymal stromal cells.88-90 MECs are enriched in expression of muscarinic and purinergic receptors.91,92 The superficial to the basement membrane location of MECs in sweat and mammary glands suggests that these cells are epithelial.93,94 Lack of vimentin expression (marker of mesenchymal cells) in MECs of salivary glands and both major and accessory LGs further supports the idea that MECs are cells of ectodermal origin.95 It has been suggested that MECs retain some proliferative potential

5

STEM CELLS IN OPHTHALMOLOGY

Figure 2 - Schematic representation of EMT and MET initiation. In healthy LGs, the acinar epithelial cells have an apico-basal polarity created by the tight junction and interactions with the basement membrane. The cells express markers of the epithelial lineage such as E-cadherin, a protein of the adherens junctions. Following injury (or inflammation), the epithelial cells undergoing EMT (steps 1 and 2) lose cell-cell and cell-extracellular matrix (ECM) contact, lose epithelial markers and express mesenchymal ones (such as vimentin), acquire front-back polarity (a morphological characteristic of migratory cells, wherein the front -leading edge- and the back show morphological and functional asymmetry) and enhanced ability to migrate. During tissue repair or resolution of inflammation (step 3), MET is initiated and the cells re-acquire their epithelial phenotype. The molecular mechanisms controlling MET are still poorly understood.

in adult uninjured LG and salivary glands, and that they have a high level of plasticity and participate in glands gland regeneration.68,96 Thus, MECs of parotid glands show a strong increase (up to 23%) in their proliferative rate 5-days following gland injury, while proliferation of other epithelial cell types (ductal and acinar) increases much later on days 7-10 after injury.96 This quick response of MECs to gland injury suggests that a MEC sub-population may contain faster proliferating committed progenitor cells.

6

Ductal cells: Similar to the duct system in the salivary glands, the LG ducts can also be divided using the same anatomical/structural divisions: intercalated, intralobular, interlobular, intralobar, interlobar, and main excretory ducts.97 Of note is the absence of striated ducts in the LG, which are commonly found in the salivary glands. LG ducts are composed of a single layer of cuboidal, eosinophilic cells that are arranged in a smooth pattern. The primary function of ductal cells is to modify the electrolyte composition of the primary LG

CHAPTER 13: Role of Stem Cells in Lacrimal and Meibomian Gland Development and Regeneration fluid (originating from the acinar cells). Several studies on regenerating salivary glands reported increased proliferation of ductal cells suggesting their involvement in tissue repair.98,99 Our own studies showed increased expression of Ki67 (a cell proliferation marker) in ductal cells of regenerating LGs.67 Furthermore, we also reported initiation of EMT within ductal cells of injured LGs suggesting the involvement of ductal cells in LG repair.52 In addition, several studies in salivary glands, as well as pancreatic and mammary glands, propose that ducts might be a niche for stem/progenitor cells capable of differentiating into other epithelial cells.100-103 Regulation of LG Development and Regeneration Our previous studies showed that LG mesenchyme plays an instructive role for epithelial cells and produces multiple growth factors that regulate LG morphogenesis and LG stem/progenitor cell specification.17,20 We, and others showed that several signaling pathways, such as FGF, Wnt and TGF-beta signaling are important for LG morphogenesis.16,17,22,104,105 For example, the FGF pathway has been shown to be particularly important for multiple aspects of embryonic development including LG epithelial bud outgrowth.16,17,106 The biological activities of FGFs and other morphogens are regulated by the ECM that establishes and maintains their gradients during development.29,64,107,108 ECM and associated growth factors are also important components of the stem cell niches.109,110 In human genetic syndromes caused by mutations of the FGF/FGFR, signaling pathway was shown to be associated with lacrimo-auriculo-dentodigital syndrome (LADD: OMIM 149730) and aplasia of lacrimal and salivary glands (ALSG:OMIM 602115). These mutations result in haplo-insufficiency of FGF10 or its receptor FGFR2b and induce severe defects in the survival and function of LG epithelial progenitor cells. Transcription factors are important regulators of tissue morphogenesis and stem cell function.111-113 We previously showed that paired-box transcription factor Pax6 is necessary for LG development and is a competence factor for LG bud outgrowth.17,20 We and others also found that the homeobox transcription factor Barx2, forkhead/winged helix transcription factor Foxc1, and Runt-related transcription factors (RUNX) are expressed in epithelial LG ducts and are necessary for normal LG development.53,114,115 Thus, autosomal dominant mutations in human FOXC1 cause eye disorders such as Axenfeld-Rieger Syndrome and glaucoma iris

hypoplasia, resulting from malformation of the anterior segment of the eye.115 More recently, we have determined that Barx2 regulates LG cell proliferation, migration and plasticity and its expression was upregulated during LG regeneration (HPM unpublished observations). LG Stem/Progenitor Cell Markers Several recent studies indicate that, similar to other exocrine glands (pancreas, salivary, and mammary),96,116-118 the LG has a high regenerative potential and that multipotent progenitor cell population(s) may be involved in their regeneration.66-68 The question that remains to be answered is whether or not LG epithelial cells have a common or specific progenitor cells. Shatos and co-authors showed that within uninjured LG nestin (an intermediate filament protein progenitor cell marker) expression was colocalized with expression of SMA and Pax6, suggesting that these MEC populations could be of epithelial origin.68 Cultured MECs expressed mesenchymal markers, such as α-actinin, vimentin and adenylyl cyclase II.92 Similar to LG, increased proliferation of MECs was described during atrophy and regeneration of rat parotid glands.96 Further studies are warranted to determine whether or not MECs could be considered as LG stem cells or differentiated cells with increased cell plasticity (i.e., that they are able to dedifferentiate and re-differentiate again). In a recent study, we have shown that Runt family transcription factors Runx1-3 are expressed in the LG epithelial component and inhibition of their expression by siRNAs blocks LG morphogenesis.114 Specifically, a high level of Runx1 expression was found within the LG ductal component and centroacinar cells. Moreover, we found that Runx1, Runx3, and cytokeratin-5 expression (putative stem cell marker) increased significantly in regenerating LGs suggesting their involvement in regulation of regeneration. Runx1 is involved in stem cell specification, proliferation and differentiation, and influences tissue homeostasis of many different organs.119-121 Runx1 was recently shown to be involved in differentiation of the epithelial secretory cell lineage in colon.122 Involvement of Runx1 in LG morphogenesis and regeneration, combined with the reported contribution of Runx1 in controlling multiple stem cell lineages, suggests that high level of Runx1 expression might mark LG epithelial progenitor cells.

7

STEM CELLS IN OPHTHALMOLOGY In addition, we reported that nestin positive cells are present in the murine lacrimal gland and that the nestin protein level increased during LG regeneration.32 We extended these findings by showing that proliferating cells within the injured murine LG were nestinpositive.66,123 These cells expressed mesenchymal, as well as progenitor cell markers such as vimentin, SMA, nestin, ABCG2 and Sca-1, and high level of proliferation as measured by Ki67 expression.66,123 In addition, a subpopulation of these cells also expressed the epithelial cell marker, Pax6, suggesting the involvement of epithelial progenitor cells in LG repair.

STRUCTURE AND DEVELOPMENT OF THE MEIBOMIAN GLANDS Because the MGs are greatly enlarged, modified sebaceous glands without a hair follicle, their structure and development is very similar to sebaceous gland morphogenesis in the skin.94 Similar to the LG, the MG is a tubuloacinar gland whose primary function is to secrete the lipid layer of the tear film.1 The MGs are embedded in the lower tarsal plates of the lid margins.75 There are approximately 30-40 glands in humans that are up to 10 mm long in the upper lid and fewer (2030) and shorter (up to 6 mm) in the lower lid.75,124 The MGs consist of a single lobule (acinus) or a collection of lobules that coalesce into a system of ducts.75,124 The duct of each gland opens directly onto the inner margin of the eyelids. Lipids are synthesized within the acinus and are then secreted into small ducts, which lead to a single, straight duct lined by modified keratinized squamous epithelium.124 MGs produce lipids that are the main component of the superficial lipid layer of the tear film. This lipid layer is thought to prevent or retard the evaporation of the aqueous layer of the tear film and to stabilize the tear film by lowering surface tension.1,75 A single meibomian gland is composed of clusters of secretory acini that are arranged circularly around a long central duct.75 The MG has holocrine secretion mode, the central duct opens at the eyelid margin, where the oily secretion is delivered onto the tear meniscus.75 MG acini can be subdivided according to their stage of differentiation and location into undifferentiated cells, maturing cells and necrotic cells.75,124 The undifferentiated cells are germinal basal cells in contact with the basement membrane and do not synthesize or secrete lipids.75,124 The maturing cells are larger and have an elaborate smooth endoplasmic reticulum and Golgi apparatus for the synthesis of lipids, which are

8

then stored in secretory granules.75,124 The necrotic cells are located in the innermost zone and are larger and full of secretory granules that are ready for release. Lipid secretion occurs through a holocrine manner: the secretory granules fuse to form large aggregates and lipid droplets and cell debris are released following lysis of the necrotic cells.75,124 Although development of sebaceous glands is well documented, little is known about MG development especially in humans.125,126 Mouse MG development starts at E18.5 with the formation of an epithelial placode within the fused eyelid margin epithelium and associated with placodes mesenchymal condensations.94 The epithelial invagination into the eyelid mesenchyme appears at P0 and at P3.5 is observed as a solid cord without lumen (Figure 3A). By P8 (Figure 3B) the developing MGs show distinct differentiation into ductal and acinar regions. By P15, the MGs appear to obtain an appearance of mature glands94 (Figure 3C-E). Mature MGs form a regular pattern in adult mouse eyelids (Figure 3F). Lipid synthesis was first detected at P3 and at P15 and it was present in the acini of the mature MG.94

MG REGENERATION AND MG STEM/PROGENITOR CELLS The information on stem cells, stem cell types and cell dynamics in the MG is very limited. Some of the currently available information on MG stem cells can be found in the review by Knop et al.127 Due to similarities with sebaceous glands, most of the information about MG stem cells is derived from studies of cell dynamics in the sebaceous glands. The holocrine type of secretion with continuous loss of acinar cells requires a mechanism that allows a continuous cell turnover and differentiation within the acinus. Several studies show that in mouse MG the rapidly dividing meibocytes are located in the basal acinar epithelium; however, these cells are most likely progenitor cells rather than stem cells, which are defined as a slow-cycling cell population. Olami and co-authors showed that meibocytes have a generation time of 4.1 days between each division.128 Interestingly, similar to other glands slow-cycling, label retaining, putative MG stem cells have been found concentrated in the ductal epithelium of the MG.129 Similarity of the MG and sebocyte suggests that MGs stem/progenitor cells may express markers found in stem/progenitor cells of sebocytes. Thus, it has been reported that sebocytes stem/progenitor cells express Blimp1 Krt5 and Krt14, while activated proliferating cells express myc, Krt5 and

CHAPTER 13: Role of Stem Cells in Lacrimal and Meibomian Gland Development and Regeneration

Figure 3 - Meibomian gland (MG) development and structure. (A) Confocal images using actin (phalloidin, red) and nuclear DNA staining of eyelids at P3.5 (postnatal day 8). At P3.5, prominent epidermal tubular-like MG invaginations can be observed (white arrows). (B) Propidium iodide nuclei staining (red) and CD31/PECAM-1 (green) staining at P8, show distinct development of acinar regions of MGs (white arrows) and blood vessels. (C and D) Hematoxylin and Eosin staining of P15 eyelids show the presence of MGs (blue dashed line) in the eyelid. (E) β-Galactosidase staining shows Runx1 expression in MGs of P15 mouse eyelids with well developed acinar region and the duct opening (red arrow, nuclei stained with fast red). Note the high level of Runx1 expression in the ductal regions and the undifferentiated meibocytes (black arrow). (F) Adult mouse eyelid showing regular patterning of fully developed MGs. Labels: hf - hair follicles, ms – muscle of eyelid.

9

STEM CELLS IN OPHTHALMOLOGY Krt14.130,131 Whether MG stem/progenitor cells express these markers has yet to be determined.

CONCLUDING REMARKS Inflammatory diseases of the lacrimal and meibomian glands that lead to tissue destruction account for the bulk of the dry eye conditions that can lead to ocular surface damage. Emerging studies from several laboratories show that both glands are capable of repair and that stem/progenitor cells play a crucial role in the repair process. Despite the modest progress, identification of lineage (epithelial and mesenchymal) specific markers for stem/progenitor cells present in the LG and MG is still lacking. Identification of these markers would therefore be an essential step toward defining the stem/ progenitor cell populations and their roles in LG and MG regeneration. Furthermore, these cells would be a great asset in the emerging field of regenerative medicine and tissue engineering, as treatment modalities to aqueousdeficient, as well as evaporative dry eye diseases. References 1.

Bron AJ, Tiffany JM, Gouveia SM, Yokoi N, Voon LW. Functional aspects of the tear film lipid layer. Exp Eye Res. 2004 Mar;78(3):347-60. 2. Dartt DA. Neural regulation of lacrimal gland secretory processes: relevance in dry eye diseases. Prog Retin Eye Res. 2009 May;28(3):155-77. 3. Tiffany JM. The normal tear film. Dev Ophthalmol. 2008;41:120. 4. Gipson IK, Argueso P. Role of mucins in the function of the corneal and conjunctival epithelia. Int Rev Cytol. 2003;231:1-49. 5. Listed NA. The definition and classification of dry eye disease: Report of the Definition and Classification Subcommittee of the International Dry Eye Workshop. Ocul Surf. 2007;Sect. 75-92. 6. Nelson JD, Shimazaki J, Benitez-del-Castillo JM, Craig JP, McCulley JP, Den S, et al. The international workshop on meibomian gland dysfunction: report of the definition and classification subcommittee. Invest Ophthalmol Vis Sci. 2011 Mar;52(4):1930-7. 7. Fox RI. Sjögren’s syndrome. Lancet. 2005 Jul 2329;366(9482):321-31. 8. Oxholm P, Asmussen K. Primary Sjögren’s syndrome: the challenge for classification of disease manifestations. J Intern Med. 1996 Jun;239(6):467-74. 9. Patel SJ, Lundy DC. Ocular manifestations of autoimmune disease. Am Fam Physician. 2002 Sep 15;66(6):991-8. 10. Schaumberg DA, Sullivan DA, Buring JE, Dana MR. Prevalence of dry eye syndrome among US women. Am J Ophthalmol. 2003 Aug;136(2):318-26. 11. Schaumberg DA, Dana R, Buring JE, Sullivan DA. Prevalence of dry eye disease among US men: estimates from the Physicians’ Health Studies. Arch Ophthalmol. 2009 Jun;127(6):763-8.

10

12. Dalzell MD. Dry eye: prevalence, utilization, and economic implications. Manag Care. 2003 Dec;12(12 Suppl):9-13. 13. Listed NA. Dry eye syndrome increases with age. Health News. 2004 May;10(5):2. 14. Moss SE, Klein R, Klein BE. Prevalence of and risk factors for dry eye syndrome. Arch Ophthalmol. 2000 Sep;118(9):1264-8. 15. Schaumberg DA, Sullivan DA, Dana MR. Epidemiology of dry eye syndrome. Adv Exp Med Biol. 2002;506(Pt B):989-98. 16. Dean C, Ito M, Makarenkova HP, Faber SC, Lang RA. Bmp7 regulates branching morphogenesis of the lacrimal gland by promoting mesenchymal proliferation and condensation. Development. 2004 Sep;131(17):4155-65. 17. Makarenkova HP, Ito M, Govindarajan V, Faber SC, Sun L, McMahon G, et al. FGF10 is an inducer and Pax6 a competence factor for lacrimal gland development. Development. 2000 Jun;127(12):2563-72. 18. Grishina IB, Makarenkova HP, Vezina CM. Bmp signaling in development and pathologies of the urethra and exocrine glands In: Nohe A, editor. Bone Morphogenetic Proteins:New Research: Nova Science Publishers, Inc.; 2012. p. 257-94. 19. Gargiulo AM, Coliolo P, Ceccarelli P, Pedini V. Ultrastructural study of sheep lacrimal glands. Veterinary research. 1999 JulAug;30(4):345-51. 20. Govindarajan V, Ito M, Makarenkova HP, Lang RA, Overbeek PA. Endogenous and ectopic gland induction by FGF-10. Dev Biol. 2000 Sep 1;225(1):188-200. 21. Lovicu FJ, Kao WW, Overbeek PA. Ectopic gland induction by lens-specific expression of keratinocyte growth factor (FGF-7) in transgenic mice. Mech Dev. 1999 Oct;88(1):43-53. 22. Entesarian M, Dahlqvist J, Shashi V, Stanley CS, Falahat B, Reardon W, et al. FGF10 missense mutations in aplasia of lacrimal and salivary glands (ALSG). Eur J Hum Genet. 2007 Mar;15(3):379-82. 23. Kalinina J, Byron SA, Makarenkova HP, Olsen SK, Eliseenkova AV, Larochelle WJ, et al. Homodimerization controls the fibroblast growth factor 9 subfamily’s receptor binding and heparan sulfate-dependent diffusion in the extracellular matrix. Mol Cell Biol. 2009 Sep;29(17):4663-78. 24. Qu X, Carbe C, Tao C, Powers A, Lawrence R, van Kuppevelt TH, et al. Lacrimal gland development and Fgf10-Fgfr2b signaling are controlled by 2-O- and 6-O-sulfated heparan sulfate. The Journal of biological chemistry. 2011 Apr 22;286(16):14435-44. 25. Fernandez-Valencia R, Gomez Pellico L. Functional anatomy of the human saccus lacrimalis. Acta anatomica. 1990;139(1):54-9. 26. de la Cuadra-Blanco C, Peces-Pena MD, Janez-Escalada L, Merida-Velasco JR. Morphogenesis of the human excretory lacrimal system. Journal of anatomy. 2006 Aug;209(2):127-35. 27. de la Cuadra-Blanco C, Peces-Pena MD, Merida-Velasco JR. Morphogenesis of the human lacrimal gland. J Anat. 2003 Nov;203(5):531-6. 28. Tripathi BJ, Tripathi RC. Evidence for the neuroectodermal origin of the human lacrimal gland. Investigative ophthalmology & visual science. 1990 Feb;31(2):393-5. 29. Makarenkova HP, Hoffman MP, Beenken A, Eliseenkova AV, Meech R, Tsau C, et al. Differential interactions of FGFs with heparan sulfate control gradient formation and branching morphogenesis. Sci Signal. 2009;2(88):ra55. 30. Williams SC, Altmann CR, Chow RL, Hemmati-Brivanlou A, Lang RA. A highly conserved lens transcriptional control element from the Pax-6 gene. Mech Dev. 1998 May;73(2):225-9.

CHAPTER 13: Role of Stem Cells in Lacrimal and Meibomian Gland Development and Regeneration 31. Kammandel B, Chowdhury K, Stoykova A, Aparicio S, Brenner S, Gruss P. Distinct cis-essential modules direct the time-space pattern of the Pax6 gene activity. Dev Biol. 1999 Jan 1;205(1):7997. 32. Zoukhri D, Fix A, Alroy J, Kublin CL. Mechanisms of murine lacrimal gland repair after experimentally induced inflammation. Invest Ophthalmol Vis Sci. 2008 Oct;49(10):4399-406. 33. Hay ED. The mesenchymal cell, its role in the embryo, and the remarkable signaling mechanisms that create it. Dev Dyn. 2005 Jul;233(3):706-20. 34. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest. 2003 Dec;112(12):177684. 35. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009 Jun;119(6):1420-8. 36. Zeisberg M, Neilson EG. Biomarkers for epithelial-mesenchymal transitions. J Clin Invest. 2009 Jun;119(6):1429-37. 37. Jechlinger M, Grunert S, Tamir IH, Janda E, Ludemann S, Waerner T, et al. Expression profiling of epithelial plasticity in tumor progression. Oncogene. 2003 Oct 16;22(46):7155-69. 38. Zavadil J, Cermak L, Soto-Nieves N, Bottinger EP. Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelialto-mesenchymal transition. The EMBO journal. 2004 Mar 10;23(5):1155-65. 39. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelialmesenchymal transitions in development and disease. Cell. 2009 Nov 25;139(5):871-90. 40. Choi SS, Omenetti A, Witek RP, Moylan CA, Syn WK, Jung Y, et al. Hedgehog pathway activation and epithelial-to-mesenchymal transitions during myofibroblastic transformation of rat hepatic cells in culture and cirrhosis. Am J Physiol Gastrointest Liver Physiol. 2009 Dec;297(6):G1093-106. 41. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008 May 16;133(4):704-15. 42. Omenetti A, Yang L, Li YX, McCall SJ, Jung Y, Sicklick JK, et al. Hedgehog-mediated mesenchymal-epithelial interactions modulate hepatic response to bile duct ligation. Lab Invest. 2007 May;87(5):499-514. 43. Sicklick JK, Choi SS, Bustamante M, McCall SJ, Perez EH, Huang J, et al. Evidence for epithelial-mesenchymal transitions in adult liver cells. Am J Physiol Gastrointest Liver Physiol. 2006 Oct;291(4):G575-83. 44. Willis BC, duBois RM, Borok Z. Epithelial origin of myofibroblasts during fibrosis in the lung. Proc Am Thorac Soc. 2006 Jun;3(4):377-82. 45. Willis BC, Liebler JM, Luby-Phelps K, Nicholson AG, Crandall ED, du Bois RM, et al. Induction of epithelial-mesenchymal transition in alveolar epithelial cells by transforming growth factor-beta1: potential role in idiopathic pulmonary fibrosis. Am J Pathol. 2005 May;166(5):1321-32. 46. Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, et al. Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci U S A. 2006 Aug 29;103(35):13180-5. 47. Battula VL, Evans KW, Hollier BG, Shi Y, Marini FC, Ayyanan A, et al. Epithelial-mesenchymal transition-derived cells exhibit multilineage differentiation potential similar to mesenchymal stem cells. Stem Cells. 2010 Aug;28(8):1435-45.

48. Morel AP, Lievre M, Thomas C, Hinkal G, Ansieau S, Puisieux A. Generation of breast cancer stem cells through epithelialmesenchymal transition. PLoS ONE. 2008;3(8):e2888. 49. Gilles C, Polette M, Zahm JM, Tournier JM, Volders L, Foidart JM, et al. Vimentin contributes to human mammary epithelial cell migration. J Cell Sci. 1999 Dec;112 ( Pt 24):4615-25. 50. Mendez MG, Kojima S, Goldman RD. Vimentin induces changes in cell shape, motility, and adhesion during the epithelial to mesenchymal transition. FASEB J. 2010 Jun;24(6):1838-51. 51. Franco DL, Mainez J, Vega S, Sancho P, Murillo MM, de Frutos CA, et al. Snail1 suppresses TGF-beta-induced apoptosis and is sufficient to trigger EMT in hepatocytes. J Cell Sci. 2010 Oct 15;123(Pt 20):3467-77. 52. You S, Avidan O, Tariq A, Ahluwalia I, Stark PC, Kublin CL, et al. Role of epithelial-mesenchymal transition in repair of the lacrimal gland after experimentally induced injury. Investigative ophthalmology & visual science. 2012 Jan;53(1):126-35. 53. Tsau C, Ito M, Gromova A, Hoffman MP, Meech R, Makarenkova HP. Barx2 and Fgf10 regulate ocular glands branching morphogenesis by controlling extracellular matrix remodeling. Development. 2011 Aug;138(15):3307-17. 54. Meech R, Edelman DB, Jones FS, Makarenkova HP. The homeobox transcription factor Barx2 regulates chondrogenesis during limb development. Development. 2005 May;132(9):213546. 55. Ader T, Norel R, Levoci L, Rogler LE. Transcriptional profiling implicates TGFbeta/BMP and Notch signaling pathways in ductular differentiation of fetal murine hepatoblasts. Mechanisms of development. 2006 Feb;123(2):177-94. 56. Ishizuya-Oka A. Epithelial-connective tissue cross-talk is essential for regeneration of intestinal epithelium. J Nippon Med Sch. 2005 Feb;72(1):13-8. 57. Grishina IB, Kim SY, Ferrara C, Makarenkova HP, Walden PD. BMP7 inhibits branching morphogenesis in the prostate gland and interferes with Notch signaling. Dev Biol. 2005 Dec 15;288(2):334-47. 58. Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes & development. 2008 Apr 1;22(7):894-907. 59. Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nature cell biology. 2008 May;10(5):593-601. 60. Tellez CS, Juri DE, Do K, Bernauer AM, Thomas CL, Damiani LA, et al. EMT and stem cell-like properties associated with miR-205 and miR-200 epigenetic silencing are early manifestations during carcinogen-induced transformation of human lung epithelial cells. Cancer research. 2011 Apr 15;71(8):3087-97. 61. Hoffmann A, Huang Y, Suetsugu-Maki R, Ringelberg CS, Tomlinson CR, Del Rio-Tsonis K, et al. Implication of the miR184 and miR-204 competitive RNA network in control of mouse secondary cataract. Molecular medicine. 2012;18:528-38. 62. Wang Y, Li W, Zang X, Chen N, Liu T, Tsonis PA, et al. MicroRNA204-5p regulates epithelial-to-mesenchymal transition during human posterior capsule opacification by targeting SMAD4. Investigative ophthalmology & visual science. 2013 Jan;54(1):323-32. 63. Pan Y, Woodbury A, Esko JD, Grobe K, Zhang X. Heparan sulfate biosynthetic gene Ndst1 is required for FGF signaling in

11

STEM CELLS IN OPHTHALMOLOGY

64.

65.

66.

67.

68.

69.

70.

71. 72.

73.

74.

75.

76.

77.

78.

79.

12

early lens development. Development. 2006 Dec;133(24):493344. Qu X, Pan Y, Carbe C, Powers A, Grobe K, Zhang X. Glycosaminoglycan-dependent restriction of FGF diffusion is necessary for lacrimal gland development. Development. 2012 Aug;139(15):2730-9. Jimenez-Rojo L, Granchi Z, Graf D, Mitsiadis TA. Stem Cell Fate Determination during Development and Regeneration of Ectodermal Organs. Frontiers in physiology. 2012;3:107. You S, Tariq A, Kublin CL, Zoukhri D. Detection of BrdU-label retaining cells in the lacrimal gland: implications for tissue repair. Cell Tissue Res. 2012 Dec;346(3):317-26. Zoukhri D, Macari E, Kublin CL. A single injection of interleukin-1 induces reversible aqueous-tear deficiency, lacrimal gland inflammation, and acinar and ductal cell proliferation. Exp Eye Res. 2007 May;84(5):894-904. Shatos MA, Haugaard-Kedstrom L, Hodges RR, Dartt DA. Isolation and characterization of progenitor cells in uninjured, adult rat lacrimal gland. Invest Ophthalmol Vis Sci. 2012 May;53(6):2749-59. Kobayashi S, Kawakita T, Kawashima M, Okada N, Mishima K, Saito I, et al. Characterization of cultivated murine lacrimal gland epithelial cells. Mol Vis.18:1271-7. Ueda Y, Karasawa Y, Satoh Y, Nishikawa S, Imaki J, Ito M. Purification and characterization of mouse lacrimal gland epithelial cells and reconstruction of an acinarlike structure in three-dimensional culture. Invest Ophthalmol Vis Sci. 2009 May;50(5):1978-87. Potten CS, Hume WJ, Reid P, Cairns J. The segregation of DNA in epithelial stem cells. Cell. 1978 Nov;15(3):899-906. Cotsarelis G, Sun TT, Lavker RM. Label-retaining cells reside in the bulge area of pilosebaceous unit: implications for follicular stem cells, hair cycle, and skin carcinogenesis. Cell. 1990 Jun 29;61(7):1329-37. Cotsarelis G, Cheng SZ, Dong G, Sun TT, Lavker RM. Existence of slow-cycling limbal epithelial basal cells that can be preferentially stimulated to proliferate: implications on epithelial stem cells. Cell. 1989 Apr 21;57(2):201-9. Davis RK, Doane MG, Knop E, Knop N, Dubielzig RR, Colitz CM, et al. Characterization of ocular gland morphology and tear composition of pinnipeds. Veterinary ophthalmology. 2012 Oct 15. Knop E, Knop N, Millar T, Obata H, Sullivan DA. The international workshop on meibomian gland dysfunction: report of the subcommittee on anatomy, physiology, and pathophysiology of the meibomian gland. Invest Ophthalmol Vis Sci. 2011;52(4):1938-78. Knop E, Knop N, Zhivov A, Kraak R, Korb DR, Blackie C, et al. The lid wiper and muco-cutaneous junction anatomy of the human eyelid margins: an in vivo confocal and histological study. J Anat. 2011 Apr;218(4):449-61. Tiwari S, Ali MJ, Balla MM, Naik MN, Honavar SG, Reddy VA, et al. Establishing human lacrimal gland cultures with secretory function. PloS one. 2012;7(1):e29458. Blackie CA, Korb DR, Knop E, Bedi R, Knop N, Holland EJ. Nonobvious obstructive meibomian gland dysfunction. Cornea. 2010 Dec;29(12):1333-45. Knop E, Korb DR, Blackie CA, Knop N. The lid margin is an underestimated structure for preservation of ocular surface health and development of dry eye disease. Developments in ophthalmology. 2010;45:108-22.

80. Knop E, Knop N, Schirra F. [Meibomian glands. Part II: physiology, characteristics, distribution and function of meibomian oil]. Der Ophthalmologe : Zeitschrift der Deutschen Ophthalmologischen Gesellschaft. [Review]. 2009 Oct;106(10):884-92. 81. Schirra F, Knop N, Knop E. [Dry eye and sex hormones: part 2]. Der Ophthalmologe : Zeitschrift der Deutschen Ophthalmologischen Gesellschaft. 2009 Nov;106(11):965. 82. Gudjonsson T, Adriance MC, Sternlicht MD, Petersen OW, Bissell MJ. Myoepithelial cells: their origin and function in breast morphogenesis and neoplasia. J Mammary Gland Biol Neoplasia. [Review ]. 2005 Jul;10(3):261-72. 83. Faraldo MM, Teuliere J, Deugnier MA, Taddei-De La Hosseraye I, Thiery JP, Glukhova MA. Myoepithelial cells in the control of mammary development and tumorigenesis: data from genetically modified mice. J Mammary Gland Biol Neoplasia. 2005 Jul;10(3):211-9. 84. Avci A, Gunhan O, Cakalagaoglu F, Gunal A, Celasun B. The cell with a thousand faces: detection of myoepithelial cells and their contributions in the cytological diagnosis of salivary gland tumors. Diagnostic cytopathology. 2012 Mar;40(3):220-7. 85. Abe J, Sugita A, Hamasaki M, Nakamura K, Iwanaga S, Nagae K, et al. Scanning electron microscopic observations of the myoepithelial cells of normal and contracting status in the rat harderian gland. The Kurume medical journal. 1981;28(2):10312. 86. Raubenheimer EJ. The myoepithelial cell: embryology, function, and proliferative aspects. Critical reviews in clinical laboratory sciences. [Review]. 1987;25(2):161-93. 87. Schon M, Benwood J, O’Connell-Willstaedt T, Rheinwald JG. Human sweat gland myoepithelial cells express a unique set of cytokeratins and reveal the potential for alternative epithelial and mesenchymal differentiation states in culture. J Cell Sci. 1999 Jun;112 (Pt 12):1925-36. 88. Wang YL, Tan Y, Satoh Y, Ono K. Morphological changes of myoepithelial cells of mouse lacrimal glands during postnatal development. Histol Histopathol. 1995 Oct;10(4):821-7. 89. Leeson TS, Leeson CR. Myoepithelial cells in the exorbital lacrimal and parotid glands of the rat in frozen-etched replicas. The American journal of anatomy. 1971 Oct;132(2):133-45. 90. Beha G, Sarli G, Brunetti B, Sassi F, Ferrara D, Benazzi C. Morphology of the myoepithelial cell: immunohistochemical characterization from resting to motile phase. Scientific World Journal. 2012;2012:252034. 91. Lemullois M, Rossignol B, Mauduit P. Immunolocalization of myoepithelial cells in isolated acini of rat exorbital lacrimal gland: cellular distribution of muscarinic receptors. Biol Cell. 1996;86(2-3):175-81. 92. Ohtomo K, Shatos MA, Vrouvlianis J, Li D, Hodges RR, Dartt DA. Increase of intracellular Ca2+ by purinergic receptors in cultured rat lacrimal gland myoepithelial cells. Investigative ophthalmology & visual science. 2011 Dec;52(13):9503-15. 93. Lin CY, Suhalim JL, Nien CL, Miljkovic MD, Diem M, Jester JV, et al. Picosecond spectral coherent anti-Stokes Raman scattering imaging with principal component analysis of meibomian glands. Journal of biomedical optics. 2011 Feb;16(2):021104. 94. Nien CJ, Massei S, Lin G, Liu H, Paugh JR, Liu CY, et al. The development of meibomian glands in mice. Mol Vis. 2010;16:1132-40. 95. Jester JV, Brown DJ. Wakayama Symposium: Peroxisome Proliferator-Activated Receptor-Gamma (PPARgamma) and Meibomian Gland Dysfunction. The ocular surface. 2012 Oct;10(4):224-9.

CHAPTER 13: Role of Stem Cells in Lacrimal and Meibomian Gland Development and Regeneration 96. Burgess KL, Dardick I, Cummins MM, Burford-Mason AP, Bassett R, Brown DH. Myoepithelial cells actively proliferate during atrophy of rat parotid gland. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 1996 Dec;82(6):674-80. 97. Ding C, Parsa L, Nandoskar P, Zhao P, Wu K, Wang Y. Duct system of the rabbit lacrimal gland: structural characteristics and role in lacrimal secretion. Invest Ophthalmol Vis Sci. 2010 Jun;51(6):2960-7. 98. Ihrler S, Blasenbreu-Vogt S, Sendelhofert A, Rossle M, Harrison JD, Lohrs U. Regeneration in chronic sialadenitis: an analysis of proliferation and apoptosis based on double immunohistochemical labelling. Virchows Arch. 2004 Apr;444(4):356-61. 99. Ihrler S, Zietz C, Sendelhofert A, Lang S, Blasenbreu-Vogt S, Lohrs U. A morphogenetic concept of salivary duct regeneration and metaplasia. Virchows Arch. 2002 May;440(5):519-26. 100. Holland AM, Gonez LJ, Harrison LC. Progenitor cells in the adult pancreas. Diabetes Metab Res Rev. 2004 Jan-Feb;20(1):13-27. 101. Zhang YQ, Kritzik M, Sarvetnick N. Identification and expansion of pancreatic stem/progenitor cells. J Cell Mol Med. 2005 AprJun;9(2):331-44. 102. Smith GH. Label-retaining epithelial cells in mouse mammary gland divide asymmetrically and retain their template DNA strands. Development. 2005 Feb;132(4):681-7. 103. Feng J, van der Zwaag M, Stokman MA, van Os R, Coppes RP. Isolation and characterization of human salivary gland cells for stem cell transplantation to reduce radiation-induced hyposalivation. Radiother Oncol. 2009 Sep;92(3):466-71. 104. Dean CH, Miller LA, Smith AN, Dufort D, Lang RA, Niswander LA. Canonical Wnt signaling negatively regulates branching morphogenesis of the lung and lacrimal gland. Dev Biol. 2005 Oct 1;286(1):270-86. 105. Pan Y, Carbe C, Powers A, Zhang EE, Esko JD, Grobe K, et al. Bud specific N-sulfation of heparan sulfate regulates Shp2dependent FGF signaling during lacrimal gland induction. Development. 2008 Jan;135(2):301-10. 106. Dean CH, Miller LA, Smith AN, Dufort D, Lang RA, Niswander LA. Canonical Wnt signaling negatively regulates branching morphogenesis of the lung and lacrimal gland. Dev Biol. 2005 Oct 1;286(1):270-86. 107. Wang J, Wang N, Xie J, Walton SC, McKown RL, Raab RW, et al. Restricted epithelial proliferation by lacritin via PKCalphadependent NFAT and mTOR pathways. J Cell Biol. 2006 Aug 28;174(5):689-700. 108. Izvolsky KI, Shoykhet D, Yang Y, Yu Q, Nugent MA, Cardoso WV. Heparan sulfate-FGF10 interactions during lung morphogenesis. Dev Biol. 2003 Jun 1;258(1):185-200. 109. Ji H, Goode RJ, Vaillant F, Mathivanan S, Kapp EA, Mathias RA, et al. Proteomic profiling of secretome and adherent plasma membranes from distinct mammary epithelial cell subpopulations. Proteomics. 2011 Oct;11(20):4029-39. 110. Schlotzer-Schrehardt U, Dietrich T, Saito K, Sorokin L, Sasaki T, Paulsson M, et al. Characterization of extracellular matrix components in the limbal epithelial stem cell compartment. Exp Eye Res. 2007 Dec;85(6):845-60. 111. Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell. 2010 May 28;38(4):576-89. 112. Collins A, Littman DR, Taniuchi I. RUNX proteins in transcription factor networks that regulate T-cell lineage choice. Nature reviews Immunology. 2009 Feb;9(2):106-15.

113. Lepper C, Fan CM. Inducible lineage tracing of Pax7-descendant cells reveals embryonic origin of adult satellite cells. Genesis. 2010 Jul;48(7):424-36. 114. Voronov D, Gromova A, Liu D, Zoukhri D, Medvinsky A, Meech R, et al. Transcription factors Runx1-3 are expressed in the lacrimal gland epithelium and are involved in regulation of gland morphogenesis and regeneration. Invest Ophthalmol Vis Sci. In press. 115. Mattiske D, Sommer P, Kidson SH, Hogan BL. The role of the forkhead transcription factor, Foxc1, in the development of the mouse lacrimal gland. Dev Dyn. 2006 Apr;235(4):1074-80. 116. Li WC, Rukstalis JM, Nishimura W, Tchipashvili V, Habener JF, Sharma A, et al. Activation of pancreatic-duct-derived progenitor cells during pancreas regeneration in adult rats. J Cell Sci. 2010 Aug 15;123(Pt 16):2792-802. 117. Lombaert IM, Knox SM, Hoffman MP. Salivary gland progenitor cell biology provides a rationale for therapeutic salivary gland regeneration. Oral Dis. [Review]. 2011 Jul;17(5):445-9. 118. Smith GH. Mammary stem cells come of age, prospectively. Trends in molecular medicine. 2006 Jul;12(7):287-9. 119. Osorio KM, Lee SE, McDermitt DJ, Waghmare SK, Zhang YV, Woo HN, et al. Runx1 modulates developmental, but not injurydriven, hair follicle stem cell activation. Development. 2008 Mar;135(6):1059-68. 120. Osorio KM, Lilja KC, Tumbar T. Runx1 modulates adult hair follicle stem cell emergence and maintenance from distinct embryonic skin compartments. J Cell Biol. 2011 Apr 4;193(1):235-50. 121. Hoi CS, Lee SE, Lu SY, McDermitt DJ, Osorio KM, Piskun CM, et al. Runx1 directly promotes proliferation of hair follicle stem cells and epithelial tumor formation in mouse skin. Mol Cell Biol. 2010 May;30(10):2518-36. 122. Fijneman RJ, Anderson RA, Richards E, Liu J, Tijssen M, Meijer GA, et al. Runx1 is a tumor suppressor gene in the mouse gastrointestinal tract. Cancer Sci. 2012 Mar;103(3):593-9. 123. You S, Kublin CL, Avidan O, Miyasaki D, Zoukhri D. Isolation and propagation of mesenchymal stem cells from the lacrimal gland. Invest Ophthalmol Vis Sci. 2011 Apr;52(5):2087-94. 124. Jester JV, Nicolaides N, Smith RE. Meibomian gland studies: histologic and ultrastructural investigations. Invest Ophthalmol Vis Sci. 1981 Apr;20(4):537-47. 125. Niemann C. Differentiation of the sebaceous gland. Dermato endocrinol. 2009 Mar;1(2):64-7. 126. Niemann C, Horsley V. Development and homeostasis of the sebaceous gland. Seminars in cell & developmental biology. 2012 Oct;23(8):928-36. 127. Knop E, Knop N, Millar T, Obata H, Sullivan DA. The international workshop on meibomian gland dysfunction: report of the subcommittee on anatomy, physiology, and pathophysiology of the meibomian gland. Invest Ophthalmol Vis Sci. [Review]. 2011 Mar;52(4):1938-78. 128. Olami Y, Zajicek G, Cogan M, Gnessin H, Pe’er J. Turnover and migration of meibomian gland cells in rats’ eyelids. Ophthalmic research. 2001 May-Jun;33(3):170-5. 129. Lavker RMT, J., Sun, T. Label-retaining Cells (LRCs) are preferentially located in the ductal epithelium of the meibomian gland: implications on the mucocutaneous junctional (MCJ) epithelium of the eyelid. Invest Ophthalmol Vis Sci. 2003;44(E-Abstract 3781). 130. Schneider MR, Paus R. Sebocytes, multifaceted epithelial cells: lipid production and holocrine secretion. The international journal of biochemistry & cell biology. 2010 Feb;42(2):181-5.

13

STEM CELLS IN OPHTHALMOLOGY 131. Horsley V, O’Carroll D, Tooze R, Ohinata Y, Saitou M, Obukhanych T, et al. Blimp1 defines a progenitor population that governs cellular input to the sebaceous gland. Cell. 2006 Aug 11;126(3):597-609.

14