November 2018
Volume 59, Issue 14
Open Access
Special Issue  |   November 2018
Advances in Functional Restoration of the Lacrimal Glands
Author Affiliations & Notes
  • Masatoshi Hirayama
    Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
    Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California, United States
  • Correspondence: Masatoshi Hirayama, Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan; [email protected]
Investigative Ophthalmology & Visual Science November 2018, Vol.59, DES174-DES182. doi:https://doi.org/10.1167/iovs.17-23528
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Masatoshi Hirayama; Advances in Functional Restoration of the Lacrimal Glands. Invest. Ophthalmol. Vis. Sci. 2018;59(14):DES174-DES182. https://doi.org/10.1167/iovs.17-23528.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
Abstract

The lacrimal glands produce tears to support a healthy homeostatic environment on the ocular surface. The lacrimal gland dysfunction characteristic of dry eye disease causes ocular discomfort and visual disturbances and in severe cases can result in a loss of vision. The demand for adequate restoration of lacrimal gland function has been intensified due to advances in stem cell biology, developmental biology, and bioengineering technologies. In addition to conventional therapies, including artificial tears, tear alternatives (such as autologous serum eye drops) and salivary gland transplantation, a regenerative medicine approach has been identified as a novel strategy to restore the function of the lacrimal gland. Recent studies have demonstrated the potential of progenitor cell injection therapy to repair the tissue of the lacrimal glands. A current three-dimensional (3D) tissue engineering technique has been shown to regenerate a secretory gland structure by reproducing reciprocal epithelial–mesenchymal interactions during ontogenesis in vitro and in vivo. A novel direct reprogramming method has suggested a possibility to induce markers in the lacrimal gland developmental process from human pluripotent stem cells. The development of this method is supported by advances in our understanding of gene expression and regulatory networks involved in the development and differentiation of the lacrimal glands. Engineering science has proposed a medical device to stimulate tearing and a bio-hybrid scaffold to reconstruct the 3D lacrimal gland structure. In this review, we will summarize recent bioengineering advances in lacrimal gland regeneration toward the functional restoration of the lacrimal glands as a future dry eye therapy.

Dry eye disease (DED) is defined at TFOS/DEWS II as a multifactorial disease of the ocular surface characterized by a loss of homeostasis of the tear film, and accompanied by ocular symptoms, in which tear film instability and hyperosmolarity, ocular surface inflammation and damage, and neurosensory abnormalities play etiological roles.1 A major role of the lacrimal glands is the maintenance of a homeostatic environment on the ocular surface epithelium through tear secretion.2,3 The functional restoration of the lacrimal glands for severe DED began with the use of alternative tear substitutes, such as artificial tear drops, to sustain a stable tear film.4 An autologous serum eye drop has been recognized as a more physiologically relevant substitute for natural tears because it contains components, such as growth factors, fibronectin, and vitamins, that support proliferation, migration and differentiation of the ocular surface epithelium.5,6 On the other hand, the transplantation of autologous accessory salivary glands into the ocular conjunctiva has been reported in the clinic as an efficient way to reconstruct the ocular surface for severe DED.7 Tear/lacrimal gland alternative therapies are some of the therapeutic choices for severe DED and have already been used in the clinic.7 Recently, regenerative medicine, which has been improved by our knowledge of developmental biology, stem cell biology, and tissue engineering technology, has arisen as a new approach to restore the function of impaired organs.8,9 In the early decades of regenerative medicine, cell injection therapies using stem cells derived from tissues and stem cell activation therapies with cytokines have been investigated to restore the function of injured organs in cases of malignant disease, myocardial infarction, and hepatic dysfunction.1012 Advances in tissue bioengineering technology allow us to generate two-dimensional tissues, including cultivated corneal epithelial cell sheets and oral mucosal epithelial cell sheets, to efficiently treat severe ocular surface diseases.1315 In addition to therapies utilizing tear/lacrimal gland substitutes, recent exponential developments in biology have advanced regenerative medicine such that reconstruction of lacrimal gland function has recently emerged as a possible method to overcome severe DED.16,17 
An organ replacement therapy based on the use of donor organs has promised a significant advantage for the restoration of organ functionality in patients with severe organ dysfunction. However, it has been difficult to solve medical issues related to donor organ transplantation, including the donor shortage and allogenic immunological rejection of the transplanted organ.1821 Developments in engineering technology have contributed to the creation of novel medical tools, such as mechanical medical devices and artificial organs, which can be used to overcome some of the problems related to donor organ transplantation.2225 One of the goals of current regenerative medicine is to replace an organ injured due to diseases, trauma, and aging with a fully functional 3D bioengineered organ created from stem cells in an ex vivo culture environment.26 Various types of technical innovation have been used to regenerate functional tissues/organs. One such innovation involves a 3D cell manipulation procedure that requires the precise arrangement of multiple kinds of cells, an optimized culture condition and a stem cell differentiation method to induce the development of bioengineered organs from available cell sources.27 
This review details the physiologic function and development of the lacrimal glands as well as recent advances in the restoration of lacrimal gland function obtained from both our works and published research. We present evidence that regenerative medicine may be utilized to make fully functional lacrimal glands in the future. 
Functional Anatomy of the Lacrimal Glands
The lacrimal glands produce an aqueous layer, which makes up the major part of the pre-corneal tear film.4 Aqueous tears contain water, electrolytes, and various secreted substances, including lipocalin and lactoferrin.28,29 Tears play indispensable physiological roles on the ocular surface, such as lid lubrication, hydration, antimicrobial activity, and protection of the ocular surface epithelium.30 
The lacrimal glands include the main lacrimal glands, which secrete the majority of the tears, and scattered small accessory lacrimal glands that exist in the ocular conjunctiva.2 The basic schematic of the lacrimal glands is that of a tubule-alveolar structure. The acini produce tear proteins and secrete fluids through aquaporin 5 (AQP5), a water channel, which is expressed on the luminal side of the cell. The fluids from the acini drain onto the ocular surface through the lacrimal gland excretory duct.2 Myoepithelial cells surrounding the acini make the acini shrink in response to nerve signals to help them to secrete tear products into the duct.31 An elaborate neural regulatory process governs lacrimal gland function in order to secrete tears.31,32 The tear-reflex loop pathway, which can secrete tears from the lacrimal glands and which depends on the stimulation of the ocular surface, is a representative example of the importance of neural regulation of tear secretion.33 The functionality of the lacrimal glands is essential for the maintenance of a physiologically normal ocular surface and visual function. 
Lacrimal Gland Development During Embryogenesis
The elaborate 3D cell distribution of secretory gland structures, including lobules of acini, ducts and myoepithelial cells, is required for efficient organ function.2 This comprehensive structure, called a tubule-alveolar structure, is formed by a developmental process during ontogenesis.34 Representative ectodermal organs, including teeth, hair follicles, salivary glands and lacrimal glands, have a common embryonic developmental strategy resulting from reciprocal epithelial and mesenchymal interactions.3537 An embryonic lacrimal gland bud, called the lacrimal gland germ, develops from the primitive ocular surface epithelium; this is followed by branching morphogenesis signaling between the embryonic epithelium and mesenchyme in the lacrimal gland bud (Figs. 1a, 1b).38 In mice, the development of the basic lacrimal gland structure is completed before birth, and the production of functional tears begins when the eye lids open.2 
Figure 1
 
Branching morphogenesis during lacrimal gland development. (a) Illustration of the differentiation and maturation of the mouse lacrimal gland. (b) Time-course microscopic observation of the development of the mouse ED16.5 lacrimal gland germ in vitro. Scale bar: 100 μm. Reprinted from Hirayama M, Ogawa M, Oshima M, et al. Functional lacrimal gland regeneration by transplantation of a bioengineered organ germ. Nat Commun. 2013;4:2497.
Figure 1
 
Branching morphogenesis during lacrimal gland development. (a) Illustration of the differentiation and maturation of the mouse lacrimal gland. (b) Time-course microscopic observation of the development of the mouse ED16.5 lacrimal gland germ in vitro. Scale bar: 100 μm. Reprinted from Hirayama M, Ogawa M, Oshima M, et al. Functional lacrimal gland regeneration by transplantation of a bioengineered organ germ. Nat Commun. 2013;4:2497.
Various kinds of fundamental signaling underlying branching morphogenesis affect the developmental regulation of the lacrimal glands (Fig. 2).16,3941 The expression of Pax6 in embryonic ocular conjunctiva leads to the onset of the lacrimal gland bud. A condensed expression of fibroblast growth factor (Fgf) 10 in the peripheral mesenchyme induces bud formation from the epithelium.38,42 The formation and outgrowth of the bud is mediated by the local activation of a signaling cascade involving N-deacetylase/N-sulfotransferase, which modifies heparin sulfate, Fgf receptors and Src homology 2-containing tyrosine phosphatase 2.42,43 Fgf signaling regulates the Sox9-Sox10 pathway, the expression of which is required for lacrimal gland branching and differentiation.44 The Fgf signaling-regulated elongation of the lacrimal gland epithelial stalk requires Barx2 expression for the control of metalloproteinases.45 The signaling between the epithelium and mesenchyme in the bud is required for the completion of branching morphogenesis.41,46 Signaling molecules in the bone morphogenetic protein (Bmp) family, including Bmp7, regulate the branching processes by managing mesenchymal proliferation and condensation.47 This series of complex biological developmental processes is critical for the creation of the histological architecture of the lacrimal glands. 
Figure 2
 
Gene expression and molecular signaling during branching morphogenesis. Elaborate gene expression and molecular signaling networks between the epithelium and mesenchyme in the lacrimal gland germ are reportedly required for the differentiation and branching morphogenesis of the lacrimal glands. One of the lacrimal gland developmental hypotheses involves the existence of a signal center (SC). ECM, extracellular matrix; HS, heparan sulfates; HSSE, heparan sulfate-synthesizing enzymes; MMPs, matrix metalloproteinases.
Figure 2
 
Gene expression and molecular signaling during branching morphogenesis. Elaborate gene expression and molecular signaling networks between the epithelium and mesenchyme in the lacrimal gland germ are reportedly required for the differentiation and branching morphogenesis of the lacrimal glands. One of the lacrimal gland developmental hypotheses involves the existence of a signal center (SC). ECM, extracellular matrix; HS, heparan sulfates; HSSE, heparan sulfate-synthesizing enzymes; MMPs, matrix metalloproteinases.
Identification of Tissue Stem/Progenitor Cells in the Lacrimal Glands for Cell Injection Therapy
Tissue stem cells, which reside in adult tissues, are responsible for maintaining organ structure and function and are activated to a proliferative state for tissue regeneration when the tissue has been partially injured.48,49 Tissue stem cells have been recognized as a clinically available candidate cell source for cell injection therapy to restore injured organ function.50,51 It has been implied that the lacrimal glands have a regenerative capacity following partial tissue injury that is mediated by tissue stem cells. This stem cell population is posited to be similar to those in other secretory glands including salivary glands, the pancreas and mammary glands, although the detailed location of the tissue stem cells in the lacrimal glands is still unknown.52 In the salivary glands—the most similar ectodermal secretory glands to the lacrimal glands—stem/progenitor cells with regenerative capabilities reportedly exist in the intercalated ducts and the basal layer of the excretory ducts.50,53 Temporary injury of the murine lacrimal glands due to interventions such as cytokine injection or excretory lacrimal gland duct ligation demonstrated that, following a temporary period of inflammation and apoptosis, tissue repair processes occur through the induction of proliferative cells expressing the stem cell markers Nestin, Ki67, ABCG2, and Sca-1.52,54 Interstitial cells with mesenchymal cell markers, subsets of cells expressing muscle-related proteins, and epithelial mesenchymal transition pathways have also been suggested as contributing to repair processes.55 Recent studies have revealed characteristics of the tissue stem/progenitor cells in the lacrimal glands. Injection of a population of epithelial progenitor cells expressing c-kit+dim/EpCAM+/Sca1/CD34/CD45 that were isolated from murine lacrimal glands were able to regenerate secretory acini in a lacrimal gland disease model.56 A small population of cells expressing cytokeratin15 (Krt15), which is commonly expressed in immature lacrimal gland epithelial cells in embryonic stages, has been observed in the basal cells of the intercalated duct in mature lacrimal glands, where the secretory gland tissue stem cells reportedly exist.57 In humans, lacrimal gland cells expressing stem cell markers such as c-kit, ABCG2 and ALDH1 have been reported.3 Research into lacrimal gland regeneration using adult tissue stem/progenitor cells has continued to advance the clinical restoration of lacrimal gland functionality. 
3D Functional Lacrimal Gland Organ Regeneration In Vivo
Development of a Novel Cell Manipulation Method for 3D Organ Regeneration
Establishment of procedures for the manipulation of several kinds of cells based on tissue bioengineering technology has been required for the generation of a 3D functional bioengineered organ.58 In addition, it is critical that the reconstructed 3D bioengineered organ be capable of coordinating with recipient tissues such as nerve and blood vessels.59 The organ germ method, which is a 3D tissue engineering technique to reconstitute a bioengineered organ germ, has emerged as a new strategy to initiate functional organ replacement regenerative therapy for severe organ dysfunction (Fig. 3a).35,60 Epithelial and mesenchymal cells, which are artificially assembled and compartmentalized at a high density using the organ germ method, were capable of reproducing the developmental process of organogenesis in vitro and building up the organ structure in a 3D collagen gel matrix.61 Bioengineered teeth and hair follicles, manipulated by the organ germ method, could successfully be engrafted and could develop into a fully functional organ with peripheral nerve coordination after transplantation.6264 We investigated the possibility of restoring lacrimal gland function by transplanting a regenerated 3D lacrimal gland organ germ created by the organ germ method.65 The bioengineered lacrimal gland organ germ, which was regenerated using ED 16.5 mouse epithelial and mesenchymal cells isolated from the lacrimal gland germ, was able to reproduce the developmental branching morphogenesis process in organ culture (Fig. 3b). The organ germ method was successfully applied to generate a bioengineered harderian gland, which produces tear lipids in mice.36,66 
Figure 3
 
3D regeneration of the bioengineered lacrimal gland using the organ germ method. (a) Conceptual illustration of 3D lacrimal gland regeneration using the organ germ method. (b) In vitro reproduction of development and branching morphogenesis in the bioengineered lacrimal gland. Scale bar: 100 μm. (c) Representative photograph of the induced duct structure in the epithelium of the bioengineered lacrimal gland (▾). Scale bar: 100 μm. (d) In vivo development of the bioengineered lacrimal gland after transplantation by the thread-guided procedure. The transplant after transplantation (▽) is shown in the left panel (Scale bar: 1 mm). The transplant at 30 days after surgery is shown in the right panel (Scale bar: 500 μm). (e) Immunohistology of the bioengineered lacrimal gland after transplantation. Aquaporin-5 (AQP5) were correctly expressed at luminal surface of the acini and duct in the left panel (AQP5, red, and E-cadherin, green). Calponin, a marker of myoepithelial cells (red) and E-cadherin (green) in the center panel, Calponin (red), neurofilament-H (NF-H; green) and DAPI (blue) in the right panel. Scale bars: 50 μm. Parts (be) reprinted from Hirayama M, Ogawa M, Oshima M, et al. Functional lacrimal gland regeneration by transplantation of a bioengineered organ germ. Nat Commun. 2013;4:2497.
Figure 3
 
3D regeneration of the bioengineered lacrimal gland using the organ germ method. (a) Conceptual illustration of 3D lacrimal gland regeneration using the organ germ method. (b) In vitro reproduction of development and branching morphogenesis in the bioengineered lacrimal gland. Scale bar: 100 μm. (c) Representative photograph of the induced duct structure in the epithelium of the bioengineered lacrimal gland (▾). Scale bar: 100 μm. (d) In vivo development of the bioengineered lacrimal gland after transplantation by the thread-guided procedure. The transplant after transplantation (▽) is shown in the left panel (Scale bar: 1 mm). The transplant at 30 days after surgery is shown in the right panel (Scale bar: 500 μm). (e) Immunohistology of the bioengineered lacrimal gland after transplantation. Aquaporin-5 (AQP5) were correctly expressed at luminal surface of the acini and duct in the left panel (AQP5, red, and E-cadherin, green). Calponin, a marker of myoepithelial cells (red) and E-cadherin (green) in the center panel, Calponin (red), neurofilament-H (NF-H; green) and DAPI (blue) in the right panel. Scale bars: 50 μm. Parts (be) reprinted from Hirayama M, Ogawa M, Oshima M, et al. Functional lacrimal gland regeneration by transplantation of a bioengineered organ germ. Nat Commun. 2013;4:2497.
Induction of a Duct Structure for Successful Engraftment of a Bioengineered Lacrimal Gland Germ
Integration of the excretory duct and the lacrimal glands lobules is essential for delivery of tear fluids to the ocular surface.2 A bioengineered lacrimal gland lacking an excretory duct structure could not continue to develop the appropriate secretory gland architecture. Our thread-guided 3D culture and transplantation technique enables the in vitro bioengineered lacrimal glands to induce the excretory duct structure from the epithelial cell aggregate and concatenate the induced duct with a recipient lacrimal gland excretory duct in a mouse with the extra-orbital lacrimal gland removed (Figs. 3c, 3d).63,65 This procedure improved the success rate of bioengineered lacrimal gland transplantation, Further, the bioengineered lacrimal gland could achieve the appropriate secretory gland architecture including a tubule-alveolar structure (i.e., lobules consisting of acini, ducts, myoepithelial cells, and peripheral nerves65; Fig. 3e). 
Restoration of Physiological Functions of the Lacrimal Glands
A primary merit of functional organ replacement therapy using 3D bioengineered organs has been the restoration of organ functions to a physiologically relevant state.8 Reflex tearing triggered via corneal thermoreceptors by a cooling stimulation on the ocular surface is a representative neural pathway for lacrimal gland function.33,67,68 The bioengineered lacrimal glands could demonstrate the reflex tearing by the cooling stimulation at the ocular surface through the host nervous system.65 This suggests that, during structural development, the bioengineered lacrimal gland germ successfully accepted host nerve innervation. 
Normal tears contain not only water but also various kinds of tear proteins, such as lactoferrin.69 These contents support physiologic tear function, including tear stability, antibacterial protection and wound repair.7072 Delivery of tear proteins and other contents of normal tears has been able to treat severe DED and diseased ocular surfaces, as demonstrated by clinical studies using tear substitutes such as autologous serum eye drops.6,7376 We demonstrated that tears secreted from the bioengineered lacrimal glands contained tear proteins, including lactoferrin, which are produced in lacrimal gland acini. These results implied that the bioengineered lacrimal gland is much more physiologically relevant in vivo than previous attempts to restore lacrimal gland function. 
One reason to work towards efficient lacrimal gland regeneration is to protect the ocular surface from DED.2 DED causes punctate keratitis (observed by fluorescein staining of the ocular surface), corneal epithelial thinning, and corneal stromal fibroblast activation.7780 We demonstrated that the ocular surface of mice engrafted with the bioengineered lacrimal gland did not show the pathologic changes characteristic of DED. The functional replacement of the lacrimal glands using 3D bioengineered lacrimal glands has demonstrated the possibility of complete restoration of lacrimal gland functionality in order to maintain a healthy ocular surface in cases of DED. 
Cutting-Edge Strategies to Regenerate the Lacrimal Glands in Humans
With the emergence of pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), technologies involving differentiation of stem cells to individual lineages have focused on the use of PSC-derived differentiated cells and tissues for regenerative medicine.81,82 A differentiation protocol using a chemically and biologically optimized culture environment has been reported for the differentiation of PSCs to target cells, such as neurons.83,84 In ophthalmology, iPSC-derived retinal pigment epithelial cells have been successfully transplanted into a patient to overcome a disease that can induce vision loss.85 A PSC differentiation technology using a self-organization process could generate biologically assembled 3D structures, such as retinal layers, and a series of ocular cell lineages, derived from SEAM.8688 The primitive ocular epithelium induced by SEAM is expected to provide a source of cells for the generation of iPSC-derived functional bioengineered lacrimal glands using the organ germ method. 
Currently, a novel strategy has been proposed to induce target cells directly from PSCs based on our understanding of transcription factor regulation of cell identity during ontogenesis.8991 One advantage of the direct conversion method is an efficient, immediate cell conversion from PSCs and somatic cells like fibroblasts to a targeted cell lineage.92 A previous report showed that the core transcription factors PAX6, OVOL2, KLF4, SOX9, TP63, and MYC could induce cells to a corneal epithelial cell lineage from human fibroblasts.93 Identification of a core transcription factor network that defines a particular cell lineage is the key to realizing the direct reprogramming of PSCs.94,95 Recently, a whole gene expression analysis based on microarray technology identified a subset of transcription factors (PAX6, FOXC1, SIX1, and SIX2) that are specifically and highly expressed in the ED16.5 mouse lacrimal gland epithelium compared with developmentally related organs such as the mature lacrimal glands, the mature/embryonic harderian glands and the embryonic ocular surface epithelium.96 These transcription factors have been implicated as part of a set of core transcription factors that define the lacrimal gland epithelial cell lineage. Specifically, the overexpression of PAX6, FOXC1, and SIX1 using transcription factor-coded synthetic mRNAs in human ESCs resulted in the differentiation of cells toward the lacrimal gland epithelial cell like phenotype accompanied by expression of a part of lacrimal gland development markers, including the immature lacrimal gland epithelial marker KRT15, the branching morphogenesis marker BARX2, and the tear secretion related markers AQP5 and lactoferrin. This result may suggest a possibility of regenerating lacrimal gland cells from human PSCs. A meta-analysis of gene expression in the human lacrimal glands has revealed the expression of specific genes and pathways in the lacrimal glands in comparison with the gene expression in other organs, such as the salivary glands, the corneal epithelium, and the conjunctiva.97 Integration of developmental biology and gene expression analyses, including next-generation sequencing technology, would provide us with new insights into lacrimal gland regeneration. 
Future Directions
The demand for restoration of lacrimal gland function using regenerative medicine for patients with severe DED has further intensified with recent advances in biology and engineering.16 Research approaches for lacrimal gland regenerative medicine have expanded from the use of tears and lacrimal gland alternatives to the induction of whole organs using stem cells (Fig. 4). Artificial lacrimal gland tear regulation via stimulation of neural pathways using medical devices has been proposed as a future therapy for DED.98 A step toward successful lacrimal gland regeneration is the establishment of technology that can construct functional secretory glands in 3D with a coherent distribution of multiple kinds of cells. The transplantation of bioengineered lacrimal gland germs has provided a proof of concept for the generation of functional bioengineered lacrimal gland structures in vivo. Previous reports demonstrated successful 3D reconstruction of the lacrimal glands using biohybrid materials, including a decellularized scaffold.99,100 The differentiation method involving self-organization of PSCs in 3D culture would be an attractive candidate for the generation of a 3D lacrimal gland organ or organoid.101 In addition, the latest bioengineering technologies, such as 3D cell printing, may contribute to the regeneration of the 3D lacrimal gland structure.102 
Figure 4
 
Current advances and future prospects in the functional restoration of the lacrimal glands. Advances in our understanding of developmental biology, stem cell biology, and engineering have improved the development of techniques for the functional restoration of the lacrimal glands. Interactive communication between conventional clinical experience, biology and engineering is required for technical innovations for future dry eye therapies.
Figure 4
 
Current advances and future prospects in the functional restoration of the lacrimal glands. Advances in our understanding of developmental biology, stem cell biology, and engineering have improved the development of techniques for the functional restoration of the lacrimal glands. Interactive communication between conventional clinical experience, biology and engineering is required for technical innovations for future dry eye therapies.
The use of available stem cell sources for the induction of lacrimal gland regeneration is one of the next challenges that must be overcome for the successful clinical application of lacrimal gland regenerative therapy.16 Cell injection therapy using tissue-derived stem cells in the lacrimal glands is nearing clinical application due to the discovery of specific cell markers that identify the epithelial progenitor cell populations in the lacrimal glands.56 Proof of the existence of such a progenitor cell in humans, an efficient culturing method for progenitor cell proliferation, the ability to efficiently obtain enough cell numbers for injection and full restoration of the damaged lacrimal glands, and investigations into optimization for human disease status would all strengthen this evidence for future clinical application. It has been shown that the immature ocular epithelium, including embryonic lacrimal gland epithelial cells, has the ability to generate a 3D lacrimal gland structure through crosstalk with mesenchymal tissues.16 The primary ocular epithelium, which is induced from iPSCs by SEAM and its downstream differentiated cell lineages, may be an available cell candidate for lacrimal gland regeneration. The direct reprogramming of PSCs has been improved upon through the progression of analyses of gene regulatory networks.103 Identification of transcription factors promoting the differentiation of lacrimal gland epithelial phenotypes from human ESCs has opened the door to the generation of human lacrimal glands. Gene expression using synthetic mRNAs is closer to clinical use than approaches using vector-based procedures because the synthetic mRNAs have the advantage of not integrating into the genome of differentiated cells.104,105 Detailed analyses of regulatory gene networks involved in lacrimal gland development are expected to result in the discovery of appropriate cell sources for the regeneration of the lacrimal glands. A future determination of core transcription factors, which can induce the development of lacrimal gland tissue from PSCs or fibroblasts, would have a potential to advance the in vivo direct reprogramming of the lacrimal glands in chronically damaged tissues.106 A multidisciplinary approach involving various areas of science, including biology and engineering, is expected to make bioengineered organ regenerative therapy a future therapy to cure DED. 
Acknowledgments
Funding of the publication fee and administration was provided by the Dry Eye Society, Tokyo, Japan. The Dry Eye Society had no role in the contents or writing of the manuscript. 
Disclosure: M. Hirayama, None 
References
Craig JP, Nichols KK, Akpek EK, et al. TFOS DEWS II definition and classification report. Ocul Surf. 2017; 15: 276– 283.
Schechter JE, Warren DW, Mircheff AK. A lacrimal gland is a lacrimal gland, but rodent's and rabbit's are not human. Ocul Surf. 2010; 8: 111– 134.
Tiwari S, Ali MJ, Balla MM, et al. Establishing human lacrimal gland cultures with secretory function. PLoS One. 2012; 7: e29458.
Mishima S. Some physiological aspects of the precorneal tear film. Arch Ophthalmol. 1965; 73: 233– 241.
Kojima T, Higuchi A, Goto E, Matsumoto Y, Dogru M, Tsubota K. Autologous serum eye drops for the treatment of dry eye diseases. Cornea. 2008; 27 (suppl 1): S25– S30.
Kojima T, Ishida R, Dogru M, et al. The effect of autologous serum eyedrops in the treatment of severe dry eye disease: a prospective randomized case-control study. Am J Ophthalmol. 2005; 139: 242– 246.
Sant' Anna AE, Hazarbassanov RM, de Freitas D, Gomes JA. Minor salivary glands and labial mucous membrane graft in the treatment of severe symblepharon and dry eye in patients with Stevens-Johnson syndrome. Br J Ophthalmol. 2012; 96: 234– 239.
Atala A. Tissue engineering, stem cells and cloning: current concepts and changing trends. Expert Opin Biol Ther. 2005; 5: 879– 892.
Brockes JP, Kumar A. Appendage regeneration in adult vertebrates and implications for regenerative medicine. Science. 2005; 310: 1919– 1923.
Copelan EA. Hematopoietic stem-cell transplantation. N Engl J Med. 2006; 354: 1813– 1826.
Segers VF, Lee RT. Stem-cell therapy for cardiac disease. Nature. 2008; 451: 937– 942.
Wang X, Foster M, Al-Dhalimy M, Lagasse E, Finegold M, Grompe M. The origin and liver repopulating capacity of murine oval cells. Proc Natl Acad Sci U S A. 2003; 100 (suppl 1): 11881– 11888.
Nishida K, Yamato M, Hayashida Y, et al. Corneal reconstruction with tissue-engineered cell sheets composed of autologous oral mucosal epithelium. N Engl J Med. 2004; 351: 1187– 1196.
Hirayama M, Satake Y, Higa K, Yamaguchi T, Shimazaki J. Transplantation of cultivated oral mucosal epithelium prepared in fibrin-coated culture dishes. Invest Ophthalmol Vis Sci. 2012; 53: 1602– 1609.
Shimazaki J, Aiba M, Goto E, Kato N, Shimmura S, Tsubota K. Transplantation of human limbal epithelium cultivated on amniotic membrane for the treatment of severe ocular surface disorders. Ophthalmology. 2002; 109: 1285– 1290.
Hirayama M, Kawakita T, Tsubota K, Shimmura S. Challenges and strategies for regenerating the lacrimal gland. Ocul Surf. 2016; 14: 135– 143.
Hirayama M, Tsubota K, Tsuji T. Functional lacrimal gland regeneration toward a next-generation organ replacement therapy. Intern Med Review. 2017. Available at: https://www.internalmedicinereview.org/index.php/imr/article/view/501.
Abouna GM. Organ shortage crisis: problems and possible solutions. Transplant Proc. 2008; 40: 34– 38.
Zakrzewski JL, van den Brink MR, Hubbell JA. Overcoming immunological barriers in regenerative medicine. Nat Biotechnol. 2014; 32: 786– 794.
Tsubota K, Satake Y, Kaido M, et al. Treatment of severe ocular-surface disorders with corneal epithelial stem-cell transplantation. N Engl J Med. 1999; 340: 1697– 1703.
Hirayama M, Yamaguchi T, Satake Y, Shimazaki J. Surgical outcome of Descemet's stripping automated endothelial keratoplasty for bullous keratopathy secondary to argon laser iridotomy. Graefes Arch Clin Exp Ophthalmol. 2012; 250: 1043– 1050.
Hirayama M, Murat D, Liu Y, Kojima T, Kawakita T, Tsubota K. Efficacy of a novel moist cool air device in office workers with dry eye disease. Acta Ophthalmol. 2013; 91: 756– 762.
Oshima M, Inoue K, Nakajima K, et al. Functional tooth restoration by next-generation bio-hybrid implant as a bio-hybrid artificial organ replacement therapy. Sci Rep. 2014; 4: 6044.
Colton CK. Implantable biohybrid artificial organs. Cell Transplant. 1995; 4: 415– 436.
Fort A, Fort N, Ricordi C, Stabler CL. Biohybrid devices and encapsulation technologies for engineering a bioartificial pancreas. Cell Transplant. 2008; 17: 997– 1003.
Nakao K, Morita R, Saji Y, et al. The development of a bioengineered organ germ method. Nat Methods. 2007; 4: 227– 230.
Ikeda E, Morita R, Nakao K, et al. Fully functional bioengineered tooth replacement as an organ replacement therapy. Proc Natl Acad Sci U S A. 2009; 106: 13475– 13480.
Dartt DA. Tear lipocalin: structure and function. Ocul Surf. 2011; 9: 126– 138.
Ohashi Y, Dogru M, Tsubota K. Laboratory findings in tear fluid analysis. Clin Chim Acta. 2006; 369: 17– 28.
Tsubota K, Satake Y, Shimazaki J. Treatment of severe dry eye. Lancet. 1996; 348: 123.
Dartt DA. Neural regulation of lacrimal gland secretory processes: relevance in dry eye diseases. Prog Retin Eye Res. 2009; 28: 155– 177.
Dartt DA. Dysfunctional neural regulation of lacrimal gland secretion and its role in the pathogenesis of dry eye syndromes. Ocul Surf. 2004; 2: 76– 91.
Belmonte C, Nichols JJ, Cox SM, et al. TFOS DEWS II pain and sensation report. Ocul Surf. 2017; 15: 404– 437.
Gomes JAP, Azar DT, Baudouin C, et al. TFOS DEWS II iatrogenic report. Ocul Surf. 2017; 15: 511– 538.
Ogawa M, Oshima M, Imamura A, et al. Functional salivary gland regeneration by transplantation of a bioengineered organ germ. Nat Commun. 2013; 4: 2498.
Hirayama M, Tsubota K, Tsuji T. Bioengineered lacrimal gland organ regeneration in vivo. J Funct Biomater. 2015; 6: 634– 649.
Oshima M, Mizuno M, Imamura A, et al. Functional tooth regeneration using a bioengineered tooth unit as a mature organ replacement regenerative therapy. PLoS One. 2011; 6: e21531.
Makarenkova HP, Ito M, Govindarajan V, et al. FGF10 is an inducer and Pax6 a competence factor for lacrimal gland development. Development. 2000; 127: 2563– 2572.
Hogan BL, Grindley J, Bellusci S, Dunn NR, Emoto H, Itoh N. Branching morphogenesis of the lung: new models for a classical problem. Cold Spring Harb Symp Quant Biol. 1997; 62: 249– 256.
Hogan BL, Yingling JM. Epithelial/mesenchymal interactions and branching morphogenesis of the lung. Curr Opin Genet Dev. 1998; 8: 481– 486.
Sakai T. Epithelial branching morphogenesis of salivary gland: exploration of new functional regulators. J Med Invest. 2009; 56 (suppl): 234– 238.
Makarenkova HP, Hoffman MP, Beenken A, et al. Differential interactions of FGFs with heparan sulfate control gradient formation and branching morphogenesis. Sci Signal. 2009; 2:ra55.
Pan Y, Carbe C, Powers A, et al. Bud specific N-sulfation of heparan sulfate regulates Shp2-dependent FGF signaling during lacrimal gland induction. Development. 2008; 135: 301– 310.
Chen Z, Huang J, Liu Y, et al. FGF signaling activates a Sox9-Sox10 pathway for the formation and branching morphogenesis of mouse ocular glands. Development. 2014; 141: 2691– 2701.
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; 138: 3307– 3317.
Rock JR, Hogan BL. Developmental biology. Branching takes nerve. Science. 2010; 329: 1610– 1611.
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; 131: 4155– 4165.
Nakada D, Levi BP, Morrison SJ. Integrating physiological regulation with stem cell and tissue homeostasis. Neuron. 2011; 70: 703– 718.
Visvader JE, Stingl J. Mammary stem cells and the differentiation hierarchy: current status and perspectives. Genes Dev. 2014; 28: 1143– 1158.
Okumura K, Shinohara M, Endo F. Capability of tissue stem cells to organize into salivary rudiments. Stem Cells Int. 2012; 2012: 502136.
Kagami H, Wang S, Hai B. Restoring the function of salivary glands. Oral Dis. 2008; 14: 15– 24.
Zoukhri D. Mechanisms involved in injury and repair of the murine lacrimal gland: role of programmed cell death and mesenchymal stem cells. Ocul Surf. 2010; 8: 60– 69.
Sumita Y, Liu Y, Khalili S, et al. Bone marrow-derived cells rescue salivary gland function in mice with head and neck irradiation. Int J Biochem Cell Biol. 2011; 43: 80– 87.
Liu Y, Hirayama M, Kawakita T, Tsubota K. A ligation of the lacrimal excretory duct in mouse induces lacrimal gland inflammation with proliferative cells. Stem Cells Int. 2017; 2017: 4923426.
Hawley D, Ding J, Thotakura S, et al. RNA-Seq and CyTOF immuno-profiling of regenerating lacrimal glands identifies a novel subset of cells expressing muscle-related proteins. PLoS One. 2017; 12: e0179385.
Gromova A, Voronov DA, Yoshida M, et al. Lacrimal gland repair using progenitor cells. Stem Cells Transl Med. 2016; 6: 88– 98.
Hirayama M, Liu Y, Kawakita T, Shimmura S, Tsubota K. Cytokeratin expression in mouse lacrimal gland germ epithelium. Exp Eye Res. 2016; 146: 54– 59.
Hirayama M, Oshima M, Tsuji T. Development and prospects of organ replacement regenerative therapy. Cornea. 2013; 32 (suppl 1): S13– S21.
Ikeda E, Tsuji T. Growing bioengineered teeth from single cells: potential for dental regenerative medicine. Expert Opin Biol Ther. 2008; 8: 735– 744.
Hirayama M. Proof of a concept for bioengineered organ replacement to restore lacrimal gland function [in Japanese]. Nippon Ganka Gakkai Zasshi. 2015; 119: 799– 806.
Hirayama M, Tsubota K, Tsuji T. Generation of a bioengineered lacrimal gland by using the organ germ method. Methods Mol Biol. 2017; 1597: 153– 165.
Asakawa K, Toyoshima KE, Ishibashi N, et al. Hair organ regeneration via the bioengineered hair follicular unit transplantation. Sci Rep. 2012; 2: 424.
Toyoshima KE, Asakawa K, Ishibashi N, et al. Fully functional hair follicle regeneration through the rearrangement of stem cells and their niches. Nature Comm. 2012; 3: 784.
Oshima M, Tsuji T. Functional tooth regenerative therapy: tooth tissue regeneration and whole-tooth replacement. Odontology. 2014; 102: 123– 136.
Hirayama M, Ogawa M, Oshima M, et al. Functional lacrimal gland regeneration by transplantation of a bioengineered organ germ. Nature Comm. 2013; 4: 2497.
Payne AP. The harderian gland: a tercentennial review. J Anat. 1994; 185 (pt 1): 1– 49.
Parra A, Madrid R, Echevarria D, et al. Ocular surface wetness is regulated by TRPM8-dependent cold thermoreceptors of the cornea. Nat Med. 2010; 16: 1396– 1399.
Robbins A, Kurose M, Winterson BJ, Meng ID. Menthol activation of corneal cool cells induces TRPM8-mediated lacrimation but not nociceptive responses in rodents. Invest Ophthalmol Vis Sci. 2012; 53: 7034– 7042.
Dilly PN. Structure and function of the tear film. Adv Exp Med Biol. 1994; 350: 239– 247.
Dursun D, Monroy D, Knighton R, et al. The effects of experimental tear film removal on corneal surface regularity and barrier function. Ophthalmology. 2000; 107: 1754– 1760.
Flanagan JL, Willcox MD. Role of lactoferrin in the tear film. Biochimie. 2009; 91: 35– 43.
Mathers WD, Lane JA, Sutphin JE, Zimmerman MB. Model for ocular tear film function. Cornea. 1996; 15: 110– 119.
Schoenwald RD, Vidvauns S, Wurster DE, Barfknecht CF. The role of tear proteins in tear film stability in the dry eye patient and in the rabbit. Adv Exp Med Biol. 1998; 438: 391– 400.
Ogawa Y, Okamoto S, Mori T, et al. Autologous serum eye drops for the treatment of severe dry eye in patients with chronic graft-versus-host disease. Bone Marrow Transplant. 2003; 31: 579– 583.
Takamura E, Shinozaki K, Hata H, Yukari J, Hori S. Efficacy of autologous serum treatment in patients with severe dry eye. Adv Exp Med Biol. 2002; 506: 1247– 1250.
Shimmura S, Ueno R, Matsumoto Y, et al. Albumin as a tear supplement in the treatment of severe dry eye. Br J Ophthalmol. 2003; 87: 1279– 1283.
Ward KW. Superficial punctate fluorescein staining of the ocular surface. Optom Vis Sci. 2008; 85: 8– 16.
Alhatem A, Cavalcanti B, Hamrah P. In vivo confocal microscopy in dry eye disease and related conditions. Semin Ophthalmol. 2012; 27: 138– 148.
Chen W, Zhang X, Liu M, et al. Trehalose protects against ocular surface disorders in experimental murine dry eye through suppression of apoptosis. Exp Eye Res. 2009; 89: 311– 318.
Xiong C, Chen D, Liu J, et al. A rabbit dry eye model induced by topical medication of a preservative benzalkonium chloride. Invest Ophthalmol Vis Sci. 2008; 49: 1850– 1856.
Okano H, Yamanaka S. iPS cell technologies: significance and applications to CNS regeneration and disease. Mol Brain. 2014; 7: 22.
Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007; 131: 861– 872.
Brennand KJ, Simone A, Jou J, et al. Modelling schizophrenia using human induced pluripotent stem cells. Nature. 2011; 473: 221– 225.
Brennand KJ, Gage FH. Concise review: the promise of human induced pluripotent stem cell-based studies of schizophrenia. Stem Cells. 2011; 29: 1915– 1922.
Mandai M, Kurimoto Y, Takahashi M. Autologous induced stem-cell-derived retinal cells for macular degeneration. N Engl J Med. 2017; 377: 792– 793.
Nakano T, Ando S, Takata N, et al. Self-formation of optic cups and storable stratified neural retina from human ESCs. Cell Stem Cell. 2012; 10: 771– 785.
Eiraku M, Takata N, Ishibashi H, et al. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature. 2011; 472: 51– 56.
Hayashi R, Ishikawa Y, Katori R, et al. Coordinated generation of multiple ocular-like cell lineages and fabrication of functional corneal epithelial cell sheets from human iPS cells. Nat Protoc. 2017; 12: 683– 696.
Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Sudhof TC, Wernig M. Direct conversion of fibroblasts to functional neurons by defined factors. Nature. 2010; 463: 1035– 1041.
Pang ZP, Yang N, Vierbuchen T, et al. Induction of human neuronal cells by defined transcription factors. Nature. 2011; 476: 220– 223.
Ieda M, Fu JD, Delgado-Olguin P, et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell. 2010; 142: 375– 386.
Yamamizu K, Piao Y, Sharov AA, et al. Identification of transcription factors for lineage-specific ESC differentiation. Stem Cell Reports. 2013; 1: 545– 559.
Kitazawa K, Hikichi T, Nakamura T, et al. OVOL2 maintains the transcriptional program of human corneal epithelium by suppressing epithelial-to-mesenchymal transition. Cell Rep. 2016; 15: 1359– 1368.
Xu J, Du Y, Deng H. Direct lineage reprogramming: strategies, mechanisms, and applications. Cell Stem Cell. 2015; 16: 119– 134.
Lee TI, Young RA. Transcriptional regulation and its misregulation in disease. Cell. 2013; 152: 1237– 1251.
Hirayama M, Ko SBH, Kawakita T, et al. Identification of transcription factors that promote the differentiation of human pluripotent stem cells into lacrimal gland epithelium-like cells. NPJ Aging Mech Dis. 2017; 3: 1.
Aakalu VK, Parameswaran S, Maienschein-Cline M, et al. Human lacrimal gland gene expression. PLoS One. 2017; 12: e0169346.
Brinton M, Kossler AL, Patel ZM, et al. Enhanced tearing by electrical stimulation of the anterior ethmoid nerve. Invest Ophthalmol Vis Sci. 2017; 58: 2341– 2348.
Spaniol K, Metzger M, Roth M, et al. Engineering of a secretory active three-dimensional lacrimal gland construct on the basis of decellularized lacrimal gland tissue. Tissue Eng Part A. 2015; 21: 2605– 2617.
Lin H, Sun G, He H, et al. Three-dimensional culture of functional adult rabbit lacrimal gland epithelial cells on decellularized scaffold. Tissue Eng Part A. 2016; 22: 65– 74.
Hynds RE, Giangreco A. Concise review: the relevance of human stem cell-derived organoid models for epithelial translational medicine. Stem Cells. 2013; 31: 417– 422.
Lee JS, Kim BS, Seo D, Park JH, Cho DW. Three-dimensional cell printing of large-volume tissues: application to ear regeneration. Tissue Eng Part C Methods. 2017; 23: 136– 145.
Ma T, Xie M, Laurent T, Ding S. Progress in the reprogramming of somatic cells. Circ Res. 2013; 112: 562– 574.
Warren L, Manos PD, Ahfeldt T, et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell. 2010; 7: 618– 630.
Mandal PK, Rossi DJ. Reprogramming human fibroblasts to pluripotency using modified mRNA. Nat Protoc. 2013; 8: 568– 582.
Sadahiro T, Yamanaka S, Ieda M. Direct cardiac reprogramming: progress and challenges in basic biology and clinical applications. Circ Res. 2015; 116: 1378– 1391.
Figure 1
 
Branching morphogenesis during lacrimal gland development. (a) Illustration of the differentiation and maturation of the mouse lacrimal gland. (b) Time-course microscopic observation of the development of the mouse ED16.5 lacrimal gland germ in vitro. Scale bar: 100 μm. Reprinted from Hirayama M, Ogawa M, Oshima M, et al. Functional lacrimal gland regeneration by transplantation of a bioengineered organ germ. Nat Commun. 2013;4:2497.
Figure 1
 
Branching morphogenesis during lacrimal gland development. (a) Illustration of the differentiation and maturation of the mouse lacrimal gland. (b) Time-course microscopic observation of the development of the mouse ED16.5 lacrimal gland germ in vitro. Scale bar: 100 μm. Reprinted from Hirayama M, Ogawa M, Oshima M, et al. Functional lacrimal gland regeneration by transplantation of a bioengineered organ germ. Nat Commun. 2013;4:2497.
Figure 2
 
Gene expression and molecular signaling during branching morphogenesis. Elaborate gene expression and molecular signaling networks between the epithelium and mesenchyme in the lacrimal gland germ are reportedly required for the differentiation and branching morphogenesis of the lacrimal glands. One of the lacrimal gland developmental hypotheses involves the existence of a signal center (SC). ECM, extracellular matrix; HS, heparan sulfates; HSSE, heparan sulfate-synthesizing enzymes; MMPs, matrix metalloproteinases.
Figure 2
 
Gene expression and molecular signaling during branching morphogenesis. Elaborate gene expression and molecular signaling networks between the epithelium and mesenchyme in the lacrimal gland germ are reportedly required for the differentiation and branching morphogenesis of the lacrimal glands. One of the lacrimal gland developmental hypotheses involves the existence of a signal center (SC). ECM, extracellular matrix; HS, heparan sulfates; HSSE, heparan sulfate-synthesizing enzymes; MMPs, matrix metalloproteinases.
Figure 3
 
3D regeneration of the bioengineered lacrimal gland using the organ germ method. (a) Conceptual illustration of 3D lacrimal gland regeneration using the organ germ method. (b) In vitro reproduction of development and branching morphogenesis in the bioengineered lacrimal gland. Scale bar: 100 μm. (c) Representative photograph of the induced duct structure in the epithelium of the bioengineered lacrimal gland (▾). Scale bar: 100 μm. (d) In vivo development of the bioengineered lacrimal gland after transplantation by the thread-guided procedure. The transplant after transplantation (▽) is shown in the left panel (Scale bar: 1 mm). The transplant at 30 days after surgery is shown in the right panel (Scale bar: 500 μm). (e) Immunohistology of the bioengineered lacrimal gland after transplantation. Aquaporin-5 (AQP5) were correctly expressed at luminal surface of the acini and duct in the left panel (AQP5, red, and E-cadherin, green). Calponin, a marker of myoepithelial cells (red) and E-cadherin (green) in the center panel, Calponin (red), neurofilament-H (NF-H; green) and DAPI (blue) in the right panel. Scale bars: 50 μm. Parts (be) reprinted from Hirayama M, Ogawa M, Oshima M, et al. Functional lacrimal gland regeneration by transplantation of a bioengineered organ germ. Nat Commun. 2013;4:2497.
Figure 3
 
3D regeneration of the bioengineered lacrimal gland using the organ germ method. (a) Conceptual illustration of 3D lacrimal gland regeneration using the organ germ method. (b) In vitro reproduction of development and branching morphogenesis in the bioengineered lacrimal gland. Scale bar: 100 μm. (c) Representative photograph of the induced duct structure in the epithelium of the bioengineered lacrimal gland (▾). Scale bar: 100 μm. (d) In vivo development of the bioengineered lacrimal gland after transplantation by the thread-guided procedure. The transplant after transplantation (▽) is shown in the left panel (Scale bar: 1 mm). The transplant at 30 days after surgery is shown in the right panel (Scale bar: 500 μm). (e) Immunohistology of the bioengineered lacrimal gland after transplantation. Aquaporin-5 (AQP5) were correctly expressed at luminal surface of the acini and duct in the left panel (AQP5, red, and E-cadherin, green). Calponin, a marker of myoepithelial cells (red) and E-cadherin (green) in the center panel, Calponin (red), neurofilament-H (NF-H; green) and DAPI (blue) in the right panel. Scale bars: 50 μm. Parts (be) reprinted from Hirayama M, Ogawa M, Oshima M, et al. Functional lacrimal gland regeneration by transplantation of a bioengineered organ germ. Nat Commun. 2013;4:2497.
Figure 4
 
Current advances and future prospects in the functional restoration of the lacrimal glands. Advances in our understanding of developmental biology, stem cell biology, and engineering have improved the development of techniques for the functional restoration of the lacrimal glands. Interactive communication between conventional clinical experience, biology and engineering is required for technical innovations for future dry eye therapies.
Figure 4
 
Current advances and future prospects in the functional restoration of the lacrimal glands. Advances in our understanding of developmental biology, stem cell biology, and engineering have improved the development of techniques for the functional restoration of the lacrimal glands. Interactive communication between conventional clinical experience, biology and engineering is required for technical innovations for future dry eye therapies.
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×