1
|
Abdal Dayem A, Bin Jang S, Lim N, Yeo HC, Kwak Y, Lee SH, Shin HJ, Cho SG. Advances in lacrimal gland organoid development: Techniques and therapeutic applications. Biomed Pharmacother 2025; 183:117870. [PMID: 39870025 DOI: 10.1016/j.biopha.2025.117870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/11/2025] [Accepted: 01/23/2025] [Indexed: 01/29/2025] Open
Abstract
The human lacrimal gland (LG), located above the outer orbital region within the frontal bone socket, is essential in maintaining eye surface health and lubrication. It is firmly anchored to the orbital periosteum by the connective tissue, and it is vital for protecting and lubricating the eye by secreting lacrimal fluid. Disruption in the production, composition, or secretion of lacrimal fluid can lead to dry eye syndrome, a condition characterized by ocular discomfort and potential eye surface damage. This review explores the recent advancements in LG organoid generation using tissues and stem cells, highlighting cutting-edge techniques in biomaterial-based and scaffold-free technologies. Additionally, we shed light on the complex pathophysiology of LG dysfunction, providing insights into the LG physiological roles while identifying strategies for generating LG organoids and exploring their potential clinical applications. Alterations in LG morphology or secretory function can affect the tear film stability and quality, leading to various ocular pathological conditions. This comprehensive review underlines the critical crosslink of LG organoid development with disease modeling and drug screening, underscoring their potential for advancing therapeutic applications.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Soo Bin Jang
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Nahee Lim
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Han Cheol Yeo
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Yeonjoo Kwak
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Shin-Hyo Lee
- Department of Anatomy, Wonkwang University School of Medicine, Iksan, Republic of Korea; Jesaeng-Euise Clinical Anatomy Center, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Hyun Jin Shin
- Konkuk University School of Medicine, Chungju city, Republic of Korea; Department of Ophthalmology, Konkuk University Medical Center, Seoul, Republic of Korea; Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea; Institute of Biomedical Science & Technology, Konkuk University, Seoul, Republic of Korea.
| | - Sang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea; R&D Team, StemExOne Co., Ltd., Seoul, Republic of Korea.
| |
Collapse
|
2
|
Takada Y, Yasuda S, Sumioka T, Okada Y, Tamura T, Yamanaka O, Saika S. Effects of Ripasudil Hydrochloride on Epithelial Repair in a Mouse Cornea. Curr Eye Res 2024; 49:1215-1222. [PMID: 39034645 DOI: 10.1080/02713683.2024.2378018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024]
Abstract
PURPOSE Effect of topical administration of a Rho kinase inhibitor, ripasudil, on epithelial wound healing in a mouse cornea was investigated. Effects of treatment of cultured human corneal epithelial cell (HCEC) line and organ-cultured corneal epithelium with ripasudil on expression of p-ERK was also examined. METHODS Epithelial defects with a diameter of 2.0 mm were prepared in the central corneas of C57BL/6 mice with or without 1-week travoprost pre-treatment, to which ripasudil or PBS as a control was instilled every 6 h immediately after preparation. The mice eyes were cultured with or without travoprost for 24-hrs. The expression levels of p-ERK in epithelium of mice eyes were compared by immunostaining after further 24-hrs culture with or without ripasudil for 24-hrs. HCEC were cultured with or without ripasudil and processed for examination for proliferation activity and protein expression of p-ERK by either immunostaining or Western blotting. The cells were also treated with or without travoprost for 24-hrs, and were further cultured with or without ripasudil. Expression levels of p-ERK were examined by Western blotting. RESULTS Ripasudil treatment suppressed post-debridement epithelial healing in association with reduced proliferation activity in peripheral (limbal) epithelium in cornea with or without pre-treatment with travoprost. Ripasudil treatment accelerated p-ERK expression. Ripasudil supplementation upregulated proliferation with increased p-ERK in HCEC. CONCLUSION Ripasudil treatment promotes wound healing of the mouse corneal epithelium by enhancing cell proliferation on peripheral (limbal) epithelium.
Collapse
Affiliation(s)
- Yukihisa Takada
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Shingo Yasuda
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Yuka Okada
- Department of Ophthalmology, Kihoku Hospital, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Tadahiko Tamura
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Osamu Yamanaka
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| |
Collapse
|
3
|
Ikeda S, Sato K, Fujita H, Ono-Minagi H, Miyaishi S, Nohno T, Ohuchi H. Harderian Gland Development and Degeneration in the Fgf10-Deficient Heterozygous Mouse. J Dev Biol 2024; 12:16. [PMID: 38921483 PMCID: PMC11205083 DOI: 10.3390/jdb12020016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
The mouse Harderian gland (HG) is a secretory gland that covers the posterior portion of the eyeball, opening at the base of the nictitating membrane. The HG serves to protect the eye surface from infection with its secretions. Mice open their eyelids at about 2 weeks of age, and the development of the HG primordium mechanically opens the eye by pushing the eyeball from its rear. Therefore, when HG formation is disturbed, the eye exhibits enophthalmos (the slit-eye phenotype), and a line of Fgf10+/- heterozygous loss-of-function mice exhibits slit-eye due to the HG atrophy. However, it has not been clarified how and when HGs degenerate and atrophy in Fgf10+/- mice. In this study, we observed the HGs in embryonic (E13.5 to E19), postnatal (P0.5 to P18) and 74-week-old Fgf10+/- mice. We found that more than half of the Fgf10+/- mice had markedly degenerated HGs, often unilaterally. The degenerated HG tissue had a melanized appearance and was replaced by connective tissue, which was observed by P10. The development of HGs was delayed or disrupted in the similar proportion of Fgf10+/- embryos, as revealed via histology and the loss of HG-marker expression. In situ hybridization showed Fgf10 expression was observed in the Harderian mesenchyme in wild-type as well as in the HG-lacking heterozygote at E19. These results show that the Fgf10 haploinsufficiency causes delayed or defective HG development, often unilaterally from the unexpectedly early neonatal period.
Collapse
Affiliation(s)
- Shiori Ikeda
- Department of Cytology and Histology, Medical School, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Keita Sato
- Department of Cytology and Histology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Hirofumi Fujita
- Department of Cytology and Histology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Hitomi Ono-Minagi
- Department of Cytology and Histology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Satoru Miyaishi
- Department of Legal Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Tsutomu Nohno
- Department of Cytology and Histology, Medical School, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Hideyo Ohuchi
- Department of Cytology and Histology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
4
|
Ibrahim N, Hifny A, Elhanbaly R, El-Desoky SMM, Gaber W. Morphogenetic events influencing corneal maturation, development, and transparency: Light and electron microscopic study. Microsc Res Tech 2023; 86:539-555. [PMID: 36695458 DOI: 10.1002/jemt.24293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/14/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023]
Abstract
The development of the cornea is a fascinating process. Its dual origin involves the differentiation of surface ectoderm cells and the migration of mesenchymal cells of neural crest origin. This research aimed to demonstrate the morphogenesis of the rabbit cornea from fetal to postnatal life using light- and electron microscopy, and immunohistochemical analysis. There were 27 rabbit embryos and nine rabbits used. The rabbit cornea begins its prenatal development on the twelfth day of gestation. The surface ectoderm differentiates into the corneal epithelium on day 13. Intriguingly, telocytes were visible within the epithelium. The secondary stroma develops on the sixteenth day of gestation by differentiation of keratocytes. At the age of 2 weeks, the lamellae of collagenous fibers become highly organized, and the stroma becomes avascular, indicating that the cornea has become transparent. Bowman's membrane appears on day 23 of pregnancy and disappears on day 30. The Descemet's membrane appears at this time and continues to thicken postnatally. The corneal endothelium appears on the twentieth gestational day as double layer of flattened cells and becomes a single layer of cuboidal cells on day 30. The spaces between the endothelial cells resemble craters. VEGF immunohistochemical expression increases over the course of development, reaching its peak in the first week after birth before decreasing in all corneal layers and becoming negative in the stroma. In conclusion, numerous morphogenetic events contribute to corneal maturation and transparency, allowing the cornea to perform its vital functions.
Collapse
Affiliation(s)
- Nagwa Ibrahim
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Abdalla Hifny
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Ruwaida Elhanbaly
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Sara M M El-Desoky
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Wafaa Gaber
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
5
|
Makrides N, Wang Q, Tao C, Schwartz S, Zhang X. Jack of all trades, master of each: the diversity of fibroblast growth factor signalling in eye development. Open Biol 2022; 12:210265. [PMID: 35016551 PMCID: PMC8753161 DOI: 10.1098/rsob.210265] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A central question in development biology is how a limited set of signalling pathways can instruct unlimited diversity of multicellular organisms. In this review, we use three ocular tissues as models of increasing complexity to present the astounding versatility of fibroblast growth factor (FGF) signalling. In the lacrimal gland, we highlight the specificity of FGF signalling in a one-dimensional model of budding morphogenesis. In the lens, we showcase the dynamics of FGF signalling in altering functional outcomes in a two-dimensional space. In the retina, we present the prolific utilization of FGF signalling from three-dimensional development to homeostasis. These examples not only shed light on the cellular basis for the perfection and complexity of ocular development, but also serve as paradigms for the diversity of FGF signalling.
Collapse
Affiliation(s)
- Neoklis Makrides
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Qian Wang
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Chenqi Tao
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Samuel Schwartz
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Xin Zhang
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University, New York, NY, USA
| |
Collapse
|
6
|
Over-expression of human PP5 gene in mice induces corneal hyperplasia and leads to ocular surface squamous neoplasia. Biochem Biophys Res Commun 2020; 529:487-493. [PMID: 32703456 DOI: 10.1016/j.bbrc.2020.06.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/05/2020] [Indexed: 01/23/2023]
Abstract
Protein phosphatase 5 (PP5) plays an important role in cell proliferation, differentiation, and development. Transgenic PP5 mice (Tg-hPP5 mice) overexpressing human PP5 gene were successfully generated by embryo injection. Tg-hPP5 mice spontaneously developed corneal hyperplasia and ocular surface squamous neoplasia (OSSN). To investigate the mechanism behind PP5-induced corneal hyperplasia, we performed immunohistochemistry, quantitative real-time PCR, and Western Blotting analyses on the corneas of Tg-hPP5 mice at 2 months and 9 months of age. We provide the first demonstration that Tg-hPP5 mice develop corneal hyperplasia at 9-months of age demonstrated via histological analysis and in vitro co-transfection investigation. We also present data that the expression of p53 is significantly reduced while the expression of FGF-7 is significantly increased in Tg-hPP5 mice with corneal hyperplasia. Co-transfection of PP5, p53, and FGF-7-promoter-driven luciferase revealed that PP5 promotes while p53 inhibits FGF-7 expression, which indicates PP5 overexpression inhibits p53 phosphorylation, thereby reducing its tumor suppressor function and increasing FGF-7 expression. In conclusion, PP5 plays a pivotal role in corneal hyperplasia development and its downregulation is a potential target for corneal hyperplasia and OSSN treatment.
Collapse
|
7
|
Molecular regulation of ocular gland development. Semin Cell Dev Biol 2018; 91:66-74. [PMID: 30266427 DOI: 10.1016/j.semcdb.2018.07.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 06/01/2018] [Accepted: 07/24/2018] [Indexed: 01/19/2023]
Abstract
The tear film is produced by two ocular glands, the lacrimal glands, which produce the aqueous component of this film, and the meibomian glands, which secrete the lipidic component that is key to reduce evaporation of the watery film at the surface of the eye. Embryonic development of these exocrine glands has been mostly studied in mice, which also develop Harderian glands, a third type of ocular gland whose role is still not well understood. This review provides an update on the signalling pathways, transcription factors andextracellular matrix components that have been shown to play a role in ocular gland development.
Collapse
|
8
|
FGF-induced Pea3 transcription factors program the genetic landscape for cell fate determination. PLoS Genet 2018; 14:e1007660. [PMID: 30188892 PMCID: PMC6143274 DOI: 10.1371/journal.pgen.1007660] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/18/2018] [Accepted: 08/27/2018] [Indexed: 12/01/2022] Open
Abstract
FGF signaling is a potent inducer of lacrimal gland development in the eye, capable of transforming the corneal epithelium into glandular tissues. Here, we show that genetic ablation of the Pea3 family of transcription factors not only disrupted the ductal elongation and branching of the lacrimal gland, but also biased the lacrimal gland epithelium toward an epidermal cell fate. Analysis of high-throughput gene expression and chromatin immunoprecipitation data revealed that the Pea3 genes directly control both the positive and negative feedback loops of FGF signaling. Importantly, Pea3 genes are also required to suppress aberrant Notch signaling which, if gone unchecked, can compromise lacrimal gland development by preventing the expression of both Sox and Six family genes. These results demonstrate that Pea3 genes are key FGF early response transcriptional factors, programing the genetic landscape for cell fate determination. FGF signaling regulates cell fate decision by inducing genome-wide changes in gene expression. We identified Pea3 family transcription factors as the key effectors of FGF signaling in reprograming the epithelia transcriptome. Pea3 factors control both the feedback and feedforward circuities of FGF signaling in lacrimal gland development. They also activate specific expression of Six and Sox family genes and suppress aberrant activation of Notch signaling. In the absence of Pea3 genes, the lacrimal gland progenitors become epidermal-like in their gene expression patterns. The study of Pea3 function resolves the long standing conundrum of how FGF induces the lacrimal gland fate, providing direction for regenerating the lacrimal gland to treat dry eye diseases.
Collapse
|
9
|
Garg A, Zhang X. Lacrimal gland development: From signaling interactions to regenerative medicine. Dev Dyn 2017; 246:970-980. [PMID: 28710815 DOI: 10.1002/dvdy.24551] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/13/2017] [Accepted: 06/06/2017] [Indexed: 12/30/2022] Open
Abstract
The lacrimal gland plays a pivotal role in keeping the ocular surface lubricated, and protecting it from environmental exposure and insult. Dysfunction of the lacrimal gland results in deficiency of the aqueous component of the tear film, which can cause dryness of the ocular surface, also known as the aqueous-deficient dry eye disease. Left untreated, this disease can lead to significant morbidity, including frequent eye infections, corneal ulcerations, and vision loss. Current therapies do not treat the underlying deficiency of the lacrimal gland, but merely provide symptomatic relief. To develop more sustainable and physiological therapies, such as in vivo lacrimal gland regeneration or bioengineered lacrimal gland implants, a thorough understanding of lacrimal gland development at the molecular level is of paramount importance. Based on the structural and functional similarities between rodent and human eye development, extensive studies have been undertaken to investigate the signaling and transcriptional mechanisms of lacrimal gland development using mouse as a model system. In this review, we describe the current understanding of the extrinsic signaling interactions and the intrinsic transcriptional network governing lacrimal gland morphogenesis, as well as recent advances in the field of regenerative medicine aimed at treating dry eye disease. Developmental Dynamics 246:970-980, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ankur Garg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, New York
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, New York
| |
Collapse
|
10
|
Sisto M, Lorusso L, Ingravallo G, Lisi S. Exocrine Gland Morphogenesis: Insights into the Role of Amphiregulin from Development to Disease. Arch Immunol Ther Exp (Warsz) 2017; 65:477-499. [DOI: 10.1007/s00005-017-0478-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 06/02/2017] [Indexed: 12/12/2022]
|
11
|
Regulating temporospatial dynamics of morphogen for structure formation of the lacrimal gland by chitosan biomaterials. Biomaterials 2016; 113:42-55. [PMID: 27810641 DOI: 10.1016/j.biomaterials.2016.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/09/2016] [Accepted: 10/11/2016] [Indexed: 11/23/2022]
Abstract
The lacrimal gland is an important organ responsible for regulating tear synthesis and secretion. The major work of lacrimal gland (LG) is to lubricate the ocular surface and maintain the health of eyes. Functional deterioration of the lacrimal gland happens because of aging, diseases, or therapeutic complications, but without effective treatments till now. The LG originates from the epithelium of ocular surface and develops by branching morphogenesis. To regenerate functional LGs, it is required to explore the way of recapitulating and facilitating the organ to establish the intricate and ramified structure. In this study, we proposed an approach using chitosan biomaterials to create a biomimetic environment beneficial to the branching structure formation of developing LG. The morphogenetic effect of chitosan was specific and optimized to promote LG branching. With chitosan, increase in temporal expression and local concentration of endogenous HGF-related molecules creates an environment around the emerging tip of LG epithelia. By efficiently enhancing downstream signaling of HGF pathways, the cellular activities and behaviors were activated to contribute to LG branching morphogenesis. The morphogenetic effect of chitosan was abolished by either ligand or receptor deprivation, or inhibition of downstream signaling transduction. Our results elucidated the underlying mechanism accounting for chitosan morphogenetic effects on LG, and also proposed promising approaches with chitosan to assist tissue structure formation of the LG.
Collapse
|
12
|
Zhang J, Upadhya D, Lu L, Reneker LW. Fibroblast growth factor receptor 2 (FGFR2) is required for corneal epithelial cell proliferation and differentiation during embryonic development. PLoS One 2015; 10:e0117089. [PMID: 25615698 PMCID: PMC4304804 DOI: 10.1371/journal.pone.0117089] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 12/19/2014] [Indexed: 11/19/2022] Open
Abstract
Fibroblast growth factors (FGFs) play important roles in many aspects of embryonic development. During eye development, the lens and corneal epithelium are derived from the same surface ectodermal tissue. FGF receptor (FGFR)-signaling is essential for lens cell differentiation and survival, but its role in corneal development has not been fully investigated. In this study, we examined the corneal defects in Fgfr2 conditional knockout mice in which Cre expression is activated at lens induction stage by Pax6 P0 promoter. The cornea in LeCre, Fgfr2loxP/loxP mice (referred as Fgfr2CKO) was analyzed to assess changes in cell proliferation, differentiation and survival. We found that Fgfr2CKO cornea was much thinner in epithelial and stromal layer when compared to WT cornea. At embryonic day 12.5–13.5 (E12.5–13.5) shortly after the lens vesicle detaches from the overlying surface ectoderm, cell proliferation (judged by labeling indices of Ki-67, BrdU and phospho-histone H3) was significantly reduced in corneal epithelium in Fgfr2CKO mice. At later stage, cell differentiation markers for corneal epithelium and underlying stromal mesenchyme, keratin-12 and keratocan respectively, were not expressed in Fgfr2CKO cornea. Furthermore, Pax6, a transcription factor essential for eye development, was not present in the Fgfr2CKO mutant corneal epithelial at E16.5 but was expressed normally at E12.5, suggesting that FGFR2-signaling is required for maintaining Pax6 expression in this tissue. Interestingly, the role of FGFR2 in corneal epithelial development is independent of ERK1/2-signaling. In contrast to the lens, FGFR2 is not required for cell survival in cornea. This study demonstrates for the first time that FGFR2 plays an essential role in controlling cell proliferation and differentiation, and maintaining Pax6 levels in corneal epithelium via ERK-independent pathways during embryonic development.
Collapse
Affiliation(s)
- Jinglin Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Dinesh Upadhya
- Dept. of Ophthalmology, Mason Eye Institute, University of Missouri, Columbia, Missouri, United States of America
| | - Lin Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Lixing W. Reneker
- Dept. of Ophthalmology, Mason Eye Institute, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
13
|
Favaron PO, Rodrigues MN, Oliveira MF, Biasi CM, Miglino MA. Embryonic and Fetal Development in - Pigmy Rice Rat - Oligoryzomys sp. (Rodentia, Sigmodontinae) and its Significance for Being a new Experimental Model. Anat Histol Embryol 2012; 41:286-99. [DOI: 10.1111/j.1439-0264.2012.01136.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2011] [Accepted: 12/01/2011] [Indexed: 11/28/2022]
Affiliation(s)
- P. O. Favaron
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science; University of Sao Paulo; Sao Paulo; SP 05508-270; Brazil
| | - M. N. Rodrigues
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science; University of Sao Paulo; Sao Paulo; SP 05508-270; Brazil
| | - M. F. Oliveira
- Department of Animal Science; Federal Rural of Semi Arid University; Mossoró; Rio Grande do Norte, 59625-900; Brazil
| | - C. M. Biasi
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science; University of Sao Paulo; Sao Paulo; SP 05508-270; Brazil
| | - M. A. Miglino
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science; University of Sao Paulo; Sao Paulo; SP 05508-270; Brazil
| |
Collapse
|
14
|
Abstract
OBJECTIVE To review the use of genetically modified mouse lines for elucidating corneal morphogenesis during embryonic development and diseases. METHODS Transgenesis and gene-targeting techniques were used to create doxycycline-inducible mouse models (tet-On) to express transgenes or ablation of LoxP-modified genes or both in corneal cells, e.g., epithelial cells, and keratocytes and periocular mesenchymal cells of neural crest origin. RESULTS Two driver mouse lines, i.e., Krt12-rtTA and Kera-rtTA, were created, which express reverse tetracycline transcription activator (rtTA) in corneal epithelial cells and keratocytes, respectively. Bitransgenic (Krt12-rtTA/tet-o-FGF7) and triple transgenic mice (Krt12rtTA/tet-o-Cre/Ctnnb1 and Kera-rtTA/tet-o-Cre/Ctnnb1) were obtained through cross-breeding tet-o-FGF7, tet-o-Cre, and Ctnnb1 mice. On doxycycline induction, overexpression of FGF7 by corneal epithelial cells of bitransgenic Krt12-rtTA/tet-o-FGF7 mice caused nuclear translocation of beta-catenin and epithelium hyperplasia resembling human ocular surface squamous neoplasia; in triple transgenic mice (Krt12rtTA/tet-o-Cre/Ctnnb1), constitutive nuclear translocation of mutant beta-catenin (loss of exon 3) leads to hyper proliferation of corneal epithelial cells; in comparison of expression of beta-catenin mutant protein by migrating, periocular mesenchymal cells of Kera-rtTA/tet-o-Cre/Ctnnb1 caused eyelid malformation. CONCLUSIONS Use of genetically modified mice is of great value to study the pathophysiology of ocular surface defects resulting from genetic mutations.
Collapse
|
15
|
Kao WWY, Liu CY. Corneal morphogenesis during development and wound healing. Jpn J Ophthalmol 2010; 54:206-10. [PMID: 20577853 DOI: 10.1007/s10384-010-0800-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 02/04/2010] [Indexed: 11/30/2022]
Affiliation(s)
- Winston W-Y Kao
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267-0838, USA.
| | | |
Collapse
|
16
|
Burgess D, Zhang Y, Siefker E, Vaca R, Kuracha MR, Reneker L, Overbeek PA, Govindarajan V. Activated Ras alters lens and corneal development through induction of distinct downstream targets. BMC DEVELOPMENTAL BIOLOGY 2010; 10:13. [PMID: 20105280 PMCID: PMC2828409 DOI: 10.1186/1471-213x-10-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 01/27/2010] [Indexed: 01/04/2023]
Abstract
Background Mammalian Ras genes regulate diverse cellular processes including proliferation and differentiation and are frequently mutated in human cancers. Tumor development in response to Ras activation varies between different tissues and the molecular basis for these variations are poorly understood. The murine lens and cornea have a common embryonic origin and arise from adjacent regions of the surface ectoderm. Activation of the fibroblast growth factor (FGF) signaling pathway induces the corneal epithelial cells to proliferate and the lens epithelial cells to exit the cell cycle. The molecular mechanisms that regulate the differential responses of these two related tissues have not been defined. We have generated transgenic mice that express a constitutively active version of human H-Ras in their lenses and corneas. Results Ras transgenic lenses and corneal epithelial cells showed increased proliferation with concomitant increases in cyclin D1 and D2 expression. This initial increase in proliferation is sustained in the cornea but not in the lens epithelial cells. Coincidentally, cdk inhibitors p27Kip1 and p57Kip2 were upregulated in the Ras transgenic lenses but not in the corneas. Phospho-Erk1 and Erk2 levels were elevated in the lens but not in the cornea and Spry 1 and Spry 2, negative regulators of Ras-Raf-Erk signaling, were upregulated more in the corneal than in the lens epithelial cells. Both lens and corneal differentiation programs were sensitive to Ras activation. Ras transgenic embryos showed a distinctive alteration in the architecture of the lens pit. Ras activation, though sufficient for upregulation of Prox1, a transcription factor critical for cell cycle exit and initiation of fiber differentiation, is not sufficient for induction of terminal fiber differentiation. Expression of Keratin 12, a marker of corneal epithelial differentiation, was reduced in the Ras transgenic corneas. Conclusions Collectively, these results suggest that Ras activation a) induces distinct sets of downstream targets in the lens and cornea resulting in distinct cellular responses and b) is sufficient for initiation but not completion of lens fiber differentiation.
Collapse
Affiliation(s)
- Daniel Burgess
- Department of Surgery, 2500 California Plaza, Creighton University, Omaha, NE 68178, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Chikama T, Liu CY, Meij JTA, Hayashi Y, Wang IJ, Yang L, Nishida T, Kao WWY. Excess FGF-7 in corneal epithelium causes corneal intraepithelial neoplasia in young mice and epithelium hyperplasia in adult mice. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:638-49. [PMID: 18276784 DOI: 10.2353/ajpath.2008.070897] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We hypothesized that human ocular surface squamous neoplasia (OSSN) may result from the continuous growth stimulation of corneal epithelial progenitor cells. In the present study, we analyzed the effects of excess fibroblast growth factor-7 (FGF-7) on both the proliferation and differentiation of corneal epithelium in a novel Krt12-rtTA/tet-O-FGF-7 double transgenic mouse model in which cornea-specific FGF-7 overexpression is achieved by doxycycline (Dox) treatment. When such adult mice were exposed to Dox, they exhibited epithelial hyperplasia with increases in phospho-extracellular signal-regulated kinase 1/2-, nuclear beta-catenin-, and 5-bromo-2'-deoxyuridine-labeled cells and altered keratin (K) 14 (K14) expression pattern, a normal K12 expression pattern, and the normal absence of K10. Hyperplasia of the adult cornea was fully reversible 2 weeks after the removal of Dox from chow. In contrast, double transgenic embryos that were exposed to Dox from embryonic day 0.5 to postnatal day 21 developed papillomatous tumors in the cornea, resembling human OSSN, and ectopic gland-like structures in the limbus, accompanied by the down-regulation of K12 and the up-regulation of K14, Pax6, and p63. These epithelial anomalies observed in young experimental mice were not fully resolved after the termination of Dox induction. Taken together, Krt12-rtTA/tet-O-FGF-7 mice may be a suitable animal model for the study of the molecular and cellular mechanisms of human OSSN.
Collapse
Affiliation(s)
- Taiichiro Chikama
- Department of Ophthalmology, University of Cincinnati Medical Center, 3223 Eden Ave., Suite 350, Cincinnati, OH 45267-0527, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Pan Y, Carbe C, Powers A, Zhang EE, Esko JD, Grobe K, Feng GS, Zhang X. Bud specific N-sulfation of heparan sulfate regulates Shp2-dependent FGF signaling during lacrimal gland induction. Development 2007; 135:301-10. [PMID: 18077586 DOI: 10.1242/dev.014829] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Preferential outgrowth of the bud cells forms the basis of branching morphogenesis. Here, we show that lacrimal gland development requires specific modification of heparan sulfates by Ndst genes at the tip of the lacrimal gland bud. Systemic and conditional knockout experiments demonstrate the tissue specific requirement of Ndst1 and Ndst2 in the lacrimal gland epithelial, but not mesenchymal, cells, and the functional importance of Ndst1 in Fgf10-Fgfr2b, but not of Fgf1-Fgfr2b, complex formation. Consistent with this, Fgf10-induced ectopic lacrimal gland budding in explant cultures is dependent upon Ndst gene dose, and epithelial deletion of Fgfr2 abolishes lacrimal gland budding, its specific modification of heparan sulfate and its phosphorylation of Shp2 (Ptpn11 - Mouse Genome Informatics). Finally, we show that genetic ablation of Ndst1, Fgfr2 or Shp2 disrupts ERK signaling in lacrimal gland budding. Given the evolutionarily conserved roles of these genes, the localized activation of the Ndst-Fgfr-Shp2 genetic cascade is probably a general regulatory mechanism of FGF signaling in branching morphogenesis.
Collapse
Affiliation(s)
- Yi Pan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Kao WWY, Liu CY. The use of transgenic and knock-out mice in the investigation of ocular surface cell biology. Ocul Surf 2007; 1:5-19. [PMID: 17075625 DOI: 10.1016/s1542-0124(12)70003-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The transgenic and knock-out mice created by transgenesis and gene targeting techniques are very useful for elucidating the pathophysiology of human diseases caused by altered genetic functions. Many of the experimental mouse lines exhibit ocular surface disorders. However, embryonic lethality and congenital defects found in many of the transgenic and knock-out mice preclude their use for studying the consequences of altered genetic functions in adult animals. To circumvent these difficulties, we have established binary inducible mouse models, using the corneal keratocyte-specific keratocan promoter, and the tetracycline-inducible gene expression system (reverse tetracycline transcription activator--rtTA). In these models, the animals function normally until they are fed doxycycline, thus inducing the overexpression of inserted transgenes by keratocytes. We have also developed inserted rtTA and Cre reporter gene constructs to create genetically modified mouse lines that have tissue-specific gene alterations to study acquired conditions, e.g., wound healing and irregular hormone and cytokine signaling that offsets homeostasis in adults. Furthermore, the genes that are ubiquitously expressed in many tissues can be specifically ablated solely in ocular surface tissues to examine their function, since the loss of such a gene in ocular surface tissues will not be life-threatening. It is noteworthy that these altered mouse lines can also be used as models for the development of therapeutic treatment regimens of diseases using gene therapy and stem cell strategies.
Collapse
Affiliation(s)
- Winston W-Y Kao
- Department of Opthalmology, University of Cincinnati, Cincinnati, OH 45267-0527, USA.
| | | |
Collapse
|
21
|
Li W, Hayashida Y, Chen YT, Tseng SCG. Niche regulation of corneal epithelial stem cells at the limbus. Cell Res 2007; 17:26-36. [PMID: 17211449 PMCID: PMC3190132 DOI: 10.1038/sj.cr.7310137] [Citation(s) in RCA: 221] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Among all adult somatic stem cells, those of the corneal epithelium are unique in their exclusive location in a defined limbal structure termed Palisades of Vogt. As a result, surgical engraftment of limbal epithelial stem cells with or without ex vivo expansion has long been practiced to restore sights in patients inflicted with limbal stem cell deficiency. Nevertheless, compared to other stem cell examples, relatively little is known about the limbal niche, which is believed to play a pivotal role in regulating self-renewal and fate decision of limbal epithelial stem cells. This review summarizes relevant literature and formulates several key questions to guide future research into better understanding of the pathogenesis of limbal stem cell deficiency and further improvement of the tissue engineering of the corneal epithelium by focusing on the limbal niche.
Collapse
Affiliation(s)
- Wei Li
- Ocular Surface Center and TissueTech Inc. Miami, Florida, 33173, USA
| | | | | | | |
Collapse
|
22
|
Abstract
Many transgenic and knockout mice exhibit pathogenic processes resembling human ocular surface diseases. Thus, the clinical manifestations of mouse lines can provide clues for identifying heritable human diseases of unknown etiology. However, mouse lines using conventional techniques of transgenesis and gene targeting often exhibit embryonic lethality and congenital defects, which preclude the use of such mouse models to study acquired ocular surface tissue diseases. These difficulties can be in part overcome by preparing mouse lines of inducible transgene expression, tissue-specific gene ablation, and inducible tissue-specific gene ablation. Conditional transgenic mouse lines live normally until administration of doxycycline and hormones that induce expression of the transgene and ablation of gene of interest. Toward this goal, we prepared 2 groups of genetically modified mouse lines: (1) transgenesis using keratocan promoter was used to create Kera-rtTA mice (doxycycline-inducible mice) and Cre-LoxP system (ie, Kera-Cre mice; conditional gene ablation in neural crest cell lineage and adult stromal keratocyte) and Kera-CrePR mice (RU-486 inducible); and (2) knock-in strategies were used to create Krt12-rtTA mice (doxycycline inducible), Krt12-Cre mice (conditional ablation in corneal epithelium), and Krt12rtTA-tet-O-Cre mice (doxycycline-inducible corneal epithelium-specific gene ablation). Using these mouse lines, we showed that transforming growth factor (TGF)-beta2 is essential for eye morphogenesis, TGF-alpha is a morphogen for eyelid formation, and lumican is a matrikine that has multiple regulatory functions on cell activities (eg, migration proliferation and gene expression) besides serving as a regulatory molecule of collagen fibrillogenesis. These mouse lines can also be used as models for development of therapeutic treatment regimens of ocular surface diseases using gene therapy and stem cell strategies.
Collapse
Affiliation(s)
- Winston W-Y Kao
- Department of Ophthalmology and Cell Biology, University of Cincinnati, 3225 Eden Avenue, Cincinnati, OH 45267, USA.
| |
Collapse
|
23
|
de la Cuadra-Blanco C, Peces-Peña MD, Jáñez-Escalada L, Mérida-Velasco JR. Morphogenesis of the human excretory lacrimal system. J Anat 2006; 209:127-35. [PMID: 16879594 PMCID: PMC2100314 DOI: 10.1111/j.1469-7580.2006.00606.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2006] [Indexed: 11/28/2022] Open
Abstract
The aim of this study was to determine the principal developmental stages in the formation of the excretory lacrimal system in humans and to establish its morphogenetic period. The study was performed using light microscopy on serial sections of 51 human specimens: 33 embryos and 18 fetuses ranging from 8 to 137 mm crown-rump length (CR; 5-16 weeks of development). Three stages were identified in the morphogenesis of the excretory lacrimal system: (1) the formative stage of the lacrimal lamina (Carnegie stages 16-18); (2) the formative stage of the lacrimal cord (Carnegie stages 19-23); and (3) the maturative stage of the excretory lacrimal system, from the 9th week of development onward. A three-dimensional reconstruction of the excretory lacrimal system was performed from serial sections of an embryo at the end of the embryonic period (27 mm CR).
Collapse
Affiliation(s)
- C de la Cuadra-Blanco
- Department of Anatomy and Embryology II, Faculty of Medicine, Complutense University of Madrid, Spain.
| | | | | | | |
Collapse
|
24
|
Finch PW, Rubin JS. Keratinocyte growth factor expression and activity in cancer: implications for use in patients with solid tumors. J Natl Cancer Inst 2006; 98:812-24. [PMID: 16788155 DOI: 10.1093/jnci/djj228] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Keratinocyte growth factor (KGF) is a locally acting epithelial mitogen that is produced by cells of mesenchymal origin and has an important role in protecting and repairing epithelial tissues. Use of recombinant human KGF (palifermin) in patients with hematologic malignancies reduces the incidence and duration of severe oral mucositis experienced after intensive chemoradiotherapy. These results suggest that KGF may be useful in the treatment of patients with other kinds of tumors, including those of epithelial origin. However, its application in this context raises issues that were not pertinent to its use in hematologic cancer because epithelial tumor cells, unlike blood cells, often express the KGF receptor (FGFR2b). Thus, it is important to examine whether KGF could promote the growth of epithelial tumors or protect such tumor cells from the effects of chemotherapy agents. Analyses of KGF and FGFR2b expression in tumor specimens and of KGF activity on transformed cells in vitro and in vivo do not indicate a definitive role for KGF in tumorigenesis. On the contrary, restoring FGFR2b expression to certain malignant cells can induce cell differentiation or apoptosis. However, other observations suggest that, in specific situations, KGF may contribute to epithelial tumorigenesis. Thus, further studies are warranted to examine the nature and extent of KGF involvement in these settings. In addition, clinical trials in patients with solid tumors are underway to assess the potential benefits of using KGF to protect normal tissue from the adverse effects of chemoradiotherapy and its possible impact on clinical outcome.
Collapse
Affiliation(s)
- Paul W Finch
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Building 37, Room 2042, 37 Convent Drive, MSC 4256, Bethesda, MD 20892-4256, USA
| | | |
Collapse
|
25
|
Carrington LM, Boulton M. Hepatocyte growth factor and keratinocyte growth factor regulation of epithelial and stromal corneal wound healing. J Cataract Refract Surg 2005; 31:412-23. [PMID: 15767167 DOI: 10.1016/j.jcrs.2004.04.072] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2004] [Indexed: 11/17/2022]
Abstract
PURPOSE To investigate the effects of hepatocyte growth factor (HGF) and keratinocyte growth factor (KGF) on early wound healing in the corneal epithelium and stroma. SETTING Cell and Molecular Biology Unit, Department of Optometry and Vision Sciences, Cardiff University, and the Cardiff Institute of Tissue Engineering and Repair, Cardiff, United Kingdom. METHODS Corneal keratocyte cell cultures and wounded corneal organ cultures (both maintained in serum-free conditions) were treated with 0.1 to 100 ng/mL of HGF or KGF for up to 5 days. Cell cultures were assessed for proliferation, migration, and differentiation into myofibroblasts. Organ cultures were used to evaluate the effect of HGF and KGF on reepithelialization following a wound, epithelial morphology and stratification, keratocyte numbers directly beneath the wounded area, and differentiation into myofibroblasts. RESULTS The 2 growth factors had opposite effects on the rate of reepithelialization, with HGF delaying and KGF accelerating epithelial coverage of the wound. Morphologic assessment showed that both growth factors affected the stratification and differentiation of the epithelium. Both factors stimulated proliferation of keratocytes in serum-free cell culture, although neither induced the appearance of myofibroblasts. This was in contrast to wounded organ cultures treated with 100 ng/mL HGF, in which large numbers of myofibroblasts were observed under the wound. Control corneas and those receiving KGF contained very few myofibroblasts. Keratocyte repopulation of the denuded area under the wound was enhanced in the presence of HGF but decreased in response to KGF. CONCLUSIONS Hepatocyte growth factor and KGF appeared to have potent and often opposite effects on epithelial and stromal cells following a wound. Hepatocyte growth factor was more detrimental than KGF, resulting in an aberrant epithelium and mass differentiation of keratocytes into myofibroblasts. Inhibition of HGF may be an appropriate therapeutic intervention in the case of persistent epithelial defects and to prevent fibrosis following a corneal stromal wound such as can occur after refractive surgery.
Collapse
Affiliation(s)
- Louise M Carrington
- Cell and Molecular Biology Unit, Department of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | | |
Collapse
|
26
|
Finch PW, Rubin JS. Keratinocyte growth factor/fibroblast growth factor 7, a homeostatic factor with therapeutic potential for epithelial protection and repair. Adv Cancer Res 2004; 91:69-136. [PMID: 15327889 DOI: 10.1016/s0065-230x(04)91003-2] [Citation(s) in RCA: 171] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Keratinocyte growth factor (KGF) is a paracrine-acting, epithelial mitogen produced by cells of mesenchymal origin. It is a member of the fibroblast growth factor (FGF) family, and acts exclusively through a subset of FGF receptor isoforms (FGFR2b) expressed predominantly by epithelial cells. The upregulation of KGF after epithelial injury suggested it had an important role in tissue repair. This hypothesis was reinforced by evidence that intestinal damage was worse and healing impaired in KGF null mice. Preclinical data from several animal models demonstrated that recombinant human KGF could enhance the regenerative capacity of epithelial tissues and protect them from a variety of toxic exposures. These beneficial effects are attributed to multiple mechanisms that collectively act to strengthen the integrity of the epithelial barrier, and include the stimulation of cell proliferation, migration, differentiation, survival, DNA repair, and induction of enzymes involved in the detoxification of reactive oxygen species. KGF is currently being evaluated in clinical trials to test its ability to ameliorate severe oral mucositis (OM) that results from cancer chemoradiotherapy. In a phase 3 trial involving patients who were treated with myeloablative chemoradiotherapy before autologous peripheral blood progenitor cell transplantation for hematologic malignancies, KGF significantly reduced both the incidence and duration of severe OM. Similar investigations are underway in patients being treated for solid tumors. On the basis of its success in ameliorating chemoradiotherapy-induced OM in humans and tissue damage in a variety of animal models, additional clinical applications of KGF are worthy of investigation.
Collapse
Affiliation(s)
- Paul W Finch
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
27
|
Dartt DA. Interaction of EGF family growth factors and neurotransmitters in regulating lacrimal gland secretion. Exp Eye Res 2004; 78:337-45. [PMID: 15106911 DOI: 10.1016/s0014-4835(03)00202-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The lacrimal gland is the primary source for the aqueous portion of the tear film. This portion contains water, electrolytes and proteins, which are necessary for the health and maintenance of the cells of the ocular surface. Afferent sensory nerves in the cornea and conjunctiva stimulate efferent parasympathetic and sympathetic nerves in the lacrimal gland. Cholinergic agonists, released from parasympathetic nevres, and norepinephrine, released from sympathetic nerves, are major stimuli of lacrimal gland secretion. These neurotransmitters activate distinct, but overlapping signal transduction pathways leading to lacrimal gland secretion. Other stimuli of lacrimal gland secretion are the EGF family of growth factors. In addition to stimulation of secretion, these growth factors can interact with the cells of the lacrimal gland themselves or with the cells of the ocular surface depending upon the location from which these growth factors are released. This review will focus on the effects of the EGF family of growth factors on the lacrimal gland and their interactions with the pathways stimulated by the neurotransmitters released from nerves.
Collapse
Affiliation(s)
- Darlene A Dartt
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA.
| |
Collapse
|
28
|
Abstract
The aim of this study was to determine the main stages of the lacrimal gland's developmental process in humans and to establish its precise morphogenetic timetable. Its onset is generally assumed to take place at O'Rahilly's stage 21, arising from an epithelial thickening of the superior extreme of the temporary conjunctival fornix. However, the present study points to a prior stage in the process: the presence of epithelial-mesenchymal changes in embryos at O'Rahilly's stage 19. The study was performed using light microscopy on serial sections of 37 human specimens: 23 embryos and 14 fetuses ranging from 15 to 137 mm crown-rump length (7-116 weeks of development). Three stages in lacrimal gland morphogenesis were identified: (1) the presumptive glandular stage, O'Rahilly's stages 19-20, characterized by a thickening of the superior fornix epithelium together with surrounding mesenchymal condensation; (2) the bud stage, generally assumed to be the first manifestation of glandular origin, characterized initially by the appearance of nodular formations in the region of the superior conjunctival fornix and concluding with the appearance of lumina within the epithelial buds; and (3) the glandular maturity stage, weeks 9-16, the period in which the gland begins to take on the morphology of adulthood.
Collapse
Affiliation(s)
- C de la Cuadra-Blanco
- Departamento de Anatomía II, Instituto de Embriología, Facultad de Medicina, Universidad Complutense, Ciudad Universitaria, Madrid, Spain.
| | | | | |
Collapse
|
29
|
Abstract
All ectodermal organs, e.g. hair, teeth, and many exocrine glands, originate from two adjacent tissue layers: the epithelium and the mesenchyme. Similar sequential and reciprocal interactions between the epithelium and mesenchyme regulate the early steps of development in all ectodermal organs. Generally, the mesenchyme provides the first instructive signal, which is followed by the formation of the epithelial placode, an early signaling center. The placode buds into or out of the mesenchyme, and subsequent proliferation, cell movements, and differentiation of the epithelium and mesenchyme contribute to morphogenesis. The molecular signals regulating organogenesis, such as molecules in the FGF, TGFbeta, Wnt, and hedgehog families, regulate the development of all ectodermal appendages repeatedly during advancing morphogenesis and differentiation. In addition, signaling by ectodysplasin, a recently identified member of the TNF family, and its receptor Edar is required for ectodermal organ development across vertebrate species. Here the current knowledge on the molecular regulation of the initiation, placode formation, and morphogenesis of ectodermal organs is discussed with emphasis on feathers, hair, and teeth.
Collapse
Affiliation(s)
- Johanna Pispa
- Developmental Biology Programme, Institute of Biotechnology, Viikki Biocenter, University of Helsinki, 00014, Helsinki, Finland
| | | |
Collapse
|
30
|
Hodges RR, Dartt DA. Regulatory pathways in lacrimal gland epithelium. INTERNATIONAL REVIEW OF CYTOLOGY 2003; 231:129-96. [PMID: 14713005 DOI: 10.1016/s0074-7696(03)31004-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tears are a complex fluid that continuously cover the exposed surface of the eye, namely the cornea and conjunctiva. Tears are secreted in response to the multitude of environmental stresses that can harm the ocular surface such as cold, mechanical stimulation, physical injury, noxious chemicals, as well as infections from various organisms. Tears also provide nutrients and remove waste from cells of the ocular surface. Because of the varied function of tears, tears are complex and are secreted by several different tissues. Tear secretion is under tight neural control allowing tears to respond rapidly to changing environmental conditions. The lacrimal gland is the main contributor to the aqueous portion of the tear film and the regulation of secretion from this gland has been well studied. Despite multiple redundencies in pathways to stimulate secretion from the lacrimal gland, defects can occur resulting in dry eye syndromes. These diseases can have deleterious effects on vision. In this review, we summarize the latest information regarding the regulatory pathways, which control secretion from the lacrimal gland, and their roles in the pathogenesis of dry eye syndromes.
Collapse
Affiliation(s)
- Robin R Hodges
- Schepens Eye Research Institute and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
31
|
Saika S, Saika S, Liu CY, Azhar M, Sanford LP, Doetschman T, Gendron RL, Kao CW, Kao WW. TGFbeta2 in corneal morphogenesis during mouse embryonic development. Dev Biol 2001; 240:419-32. [PMID: 11784073 DOI: 10.1006/dbio.2001.0480] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To examine the roles of TGFbeta isoforms on corneal morphogenesis, the eyes of mice that lack TGFbetas were analyzed at different developmental stages for cell proliferation, migration and apoptosis, and for expression patterns of keratin 12, lumican, keratocan and collagen I. Among the three Tgfb(-/-) mice, only Tgfb2(-/-) mice have abnormal ocular morphogenesis characterized by thin corneal stroma, absence of corneal endothelium, fusion of cornea to lens (a Peters'-like anomaly phenotype), and accumulation of hyaline cells in vitreous. In Tgfb2(-/-) mice, fewer keratocytes were found in stroma that has a decreased accumulation of ECM; for example, lumican, keratocan and collagen I were greatly diminished. The absence of TGFbeta2 did not compromise cell proliferation, nor enhance apoptosis. The thinner stroma resulting from decreased ECM synthesis may account for the decreased cell number in the stroma of Tgfb2 null mice. Keratin 12 expression was not altered in Tgfb2(-/-) mice, implicating normal corneal type epithelial differentiation. Delayed appearance of macrophages in ocular tissues was observed in Tgfb2(-/-) mice. Malfunctioning macrophages may account for accumulation of cell mass in vitreous of Tgfb2 null mice.
Collapse
Affiliation(s)
- S Saika
- Department of Ophthalmology, University of Cincinnati, Cincinnati, Ohio, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
One of the important functions of the cornea is to maintain normal vision by refracting light onto the lens and retina. This property is dependent in part on the ability of the corneal epithelium to undergo continuous renewal. Epithelial renewal is essential because it enables this tissue to act as a barrier that protects the corneal interior from becoming infected by noxious environmental agents. Furthermore, the smooth optical properties of the corneal epithelial surface are sustained through this renewal process. The rate of renewal is dependent on a highly integrated balance between the processes of corneal epithelial proliferation, differentiation, and cell death. One experimental approach to characterize these three aspects of the renewal process has been to study the kinetics and dynamics of corneal re-epithelialization in a wound-healing model. This effort has employed in vivo and in vitro studies. From such studies it is evident that the appropriate integration and coordination of corneal epithelial proliferation, adhesion, migration, and cell demise is dependent on the actions of a myriad of cytokines. Our goal here is to provide an overview into how these mediators and environmental factors elicit control of cellular proliferation, adhesion, migration, and apoptosis. To this end we review the pertinent literature dealing with the receptor and the cell signaling events that are responsible for mediating cytokine control of corneal epithelial renewal. It is our hope that a better appreciation can be obtained about the complexity of the control processes that are responsible for assuring continuous corneal epithelial renewal in health and disease.
Collapse
Affiliation(s)
- L Lu
- Department of Physiology and Biophysics, Wright State University School of Medicine, Dayton, OH 45435, USA.
| | | | | |
Collapse
|
33
|
Abstract
The vertebrate lens has a distinct polarity with cuboidal epithelial cells on the anterior side and differentiated fiber cells on the posterior side. It has been proposed that the anterior-posterior polarity of the lens is imposed by factors present in the ocular media surrounding the lens (aqueous and vitreous humor). The differentiation factors have been hypothesized to be members of the fibroblast growth factor (FGF) family. Though FGFs have been shown to be sufficient for induction of lens differentiation both in vivo and in vitro, they have not been demonstrated to be necessary for endogenous initiation of fiber cell differentiation. To test this possibility, we have generated transgenic mice with ocular expression of secreted self-dimerizing versions of FGFR1 (FR1) and FGFR3 (FR3). Expression of FR3, but not FR1, leads to an expansion of proliferating epithelial cells from the anterior to the posterior side of the lens due to a delay in the initiation of fiber cell differentiation. This delay is most apparent postnatally and correlates with appropriate changes in expression of marker genes including p57(KIP2), Maf and Prox1. Phosphorylation of Erk1 and Erk2 was reduced in the lenses of FR3 mice compared with nontransgenic mice. Though differentiation was delayed in FR3 mice, the lens epithelial cells still retained their intrinsic ability to respond to FGF stimulation. Based on these results we propose that the initiation of lens fiber cell differentiation in mice requires FGF receptor signaling and that one of the lens differentiation signals in the vitreous humor is a ligand for FR3, and is therefore likely to be an FGF or FGF-like factor.
Collapse
MESH Headings
- Animals
- Body Patterning
- Cell Differentiation
- Crystallins/biosynthesis
- Cyclin-Dependent Kinase Inhibitor p57
- DNA-Binding Proteins/isolation & purification
- Embryonic Induction
- Epithelial Cells
- Fibroblast Growth Factors
- Homeodomain Proteins/isolation & purification
- Lens, Crystalline/blood supply
- Lens, Crystalline/cytology
- Lens, Crystalline/embryology
- Mice
- Mice, Transgenic
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3
- Mitogen-Activated Protein Kinases/metabolism
- Models, Biological
- Nuclear Proteins/isolation & purification
- Phosphorylation
- Protein-Tyrosine Kinases
- Proto-Oncogene Proteins/isolation & purification
- Proto-Oncogene Proteins c-maf
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Fibroblast Growth Factor, Type 1
- Receptor, Fibroblast Growth Factor, Type 3
- Receptors, Fibroblast Growth Factor/metabolism
- Transgenes
- Tumor Suppressor Proteins
Collapse
Affiliation(s)
- V Govindarajan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
34
|
Kurpakus-Wheater M, Kernacki KA, Hazlett LD. Maintaining Corneal Integrity How the “Window” Stays Clear. ACTA ACUST UNITED AC 2001. [DOI: 10.1016/s0079-6336(01)80003-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
35
|
Govindarajan V, Ito M, Makarenkova HP, Lang RA, Overbeek PA. Endogenous and ectopic gland induction by FGF-10. Dev Biol 2000; 225:188-200. [PMID: 10964474 DOI: 10.1006/dbio.2000.9812] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
FGF-10, a member of the fibroblast growth factor family, is expressed in mesodermally derived cell populations during embryogenesis. During normal ocular development, FGF-10 is expressed in the perioptic mesenchyme adjacent to the Harderian and lacrimal gland primordia. In this report, we provide evidence that FGF-10 is both necessary and sufficient to initiate glandular morphogenesis. Lens-specific expression of FGF-10 was sufficient to induce ectopic ocular glands within the cornea. In addition, lacrimal and Harderian glands were not seen in FGF-10 null fetuses. Based on these results we propose that FGF-10 is an inductive signal that initiates ocular gland morphogenesis.
Collapse
Affiliation(s)
- V Govindarajan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, 77030, USA
| | | | | | | | | |
Collapse
|
36
|
Makarenkova HP, Ito M, Govindarajan V, Faber SC, Sun L, McMahon G, Overbeek PA, Lang RA. FGF10 is an inducer and Pax6 a competence factor for lacrimal gland development. Development 2000; 127:2563-72. [PMID: 10821755 DOI: 10.1242/dev.127.12.2563] [Citation(s) in RCA: 153] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We investigated the mechanism of tissue induction and specification using the lacrimal gland as a model system. This structure begins its morphogenesis as a bud-like outgrowth of the conjunctival epithelium and ultimately forms a branched structure with secretory function. Using a reporter transgene as a specific marker for gland epithelium, we show that the transcription factor Pax6 is required for normal development of the gland and is probably an important competence factor. In investigating the cell-cell signaling required, we show that fibroblast growth factor (FGF) 10 is sufficient to stimulate ectopic lacrimal bud formation in ocular explants. Expression of FGF10 in the mesenchyme adjacent to the presumptive lacrimal bud and absence of lacrimal gland development in FGF10-null mice strongly suggest that it is an endogenous inducer. This was supported by the observation that inhibition of signaling by a receptor for FGF10 (receptor 2 IIIb) suppressed development of the endogenous lacrimal bud. In explants of mesenchyme-free gland epithelium, FGF10 stimulated growth but not branching morphogenesis. This suggested that its role in induction is to stimulate proliferation and, in turn, that FGF10 combines with other factors to provide the instructive signals required for lacrimal gland development.
Collapse
Affiliation(s)
- H P Makarenkova
- Skirball Institute for Biomolecular Medicine, Developmental Genetics Program, Cell Biology and Pathology Departments, New York University Medical Center, New York, NY 10016, USA
| | | | | | | | | | | | | | | |
Collapse
|