Dry eye disease (DED) is a chronic immune-mediated disorder of the ocular surface, characterized by disruption of the epithelial barrier and sustained ocular surface inflammation, which in severe cases results in blindness from corneal ulceration and scarring. DED is arguably the most common ophthalmologic condition for which patients visit eye care professionals, with more than 16 million US adults estimated to have been diagnosed with DED (several folds higher than age-related macular degeneration). Notably, the prevalence of DED increases with age. The financial burden of DED to the US healthcare system is vast, with direct costs and expenses from productivity losses estimated to exceed $3.5 billion annually. The work of several laboratories, including the Principal Investigator's, has revealed key insights concerning the immunopathogenesis of DED. There is now strong evidence that T helper-17 (Th17) cells are critical promoters of immune-mediated damage to the ocular surface. The PI's laboratory has shown that memory Th17 (mTh17) cells maintain disease chronicity, resulting in ocular surface epitheliopathy that persists for many months after exposure to desiccating stress. Moreover, data from the PI's lab show that regulatory T cells (Tregs) are defective in suppressing the Th17 response in DED. In addition, the PI's lab has identified critical mechanisms controlling antigen-presenting cell (APC) activation and trafficking in DED, as well as effector Th17 priming, expansion and homing. Despite substantial progress in unraveling underlying immunopathogenic mechanisms, important questions remain unanswered. We hypothesize that (1) chronic DED is characterized by long-term impairment in the function of regulatory T cells (Tregs) that normally maintain immune quiescence, and that (2) the aged are more susceptible to DED due to a larger pool of memory Th17, whose function is inadequately regulated by aged Tregs. The principal objectives of this project are to (i) precisely define the phenotypic changes that characterize, and the mechanisms that promote long-term Treg dysfunction in DED; (ii) determine the cellular precursors of mTh17 cells, as well as the cytokine mechanisms that influence the generation of immunologic memory; and (iii) define the factors that amplify mTh17 immunity in aging. To achieve these objectives, three specific aims have been defined to answer the following questions:
Aim 1 : What are the causative mechanisms of enduring Treg dysfunction in DED? Aim 2: Which effector Th17 subsets are predisposed to enter the memory pool (thus permitting DED chronicity), and what factors regulate this process? And finally Aim 3: What are the cellular and molecular mechanisms that augment Th17-mediated DED in aging? It is anticipated that this research will have significant translational impact given the high prevalence of DED, the still limited knowledge we have regarding its immunopathogenesis, and the relative scarcity of effective treatments.

Public Health Relevance

Dry eye disease is a highly prevalent inflammatory disorder of the ocular surface. Associated with visual dysfunction, discomfort, and a heightened risk of corneal scarring and ulceration, the immunopathogenic mechanisms of this common condition are as of yet incompletely understood. We propose to determine the specific immune-based cellular and molecular pathogenic elements of dry eye disease, anticipating that improved understanding of these pathogenic mechanisms will yield therapeutic targets that could be useful in optimizing the management of the millions of Americans suffering from dry eye disease.

Agency
National Institute of Health (NIH)
Institute
National Eye Institute (NEI)
Type
Research Project (R01)
Project #
5R01EY020889-11
Application #
10063525
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Mckie, George Ann
Project Start
2010-09-30
Project End
2024-11-30
Budget Start
2020-12-01
Budget End
2021-11-30
Support Year
11
Fiscal Year
2021
Total Cost
Indirect Cost
Name
Schepens Eye Research Institute
Department
Type
DUNS #
073826000
City
Boston
State
MA
Country
United States
Zip Code
02114
Tan, Xuhua; Chen, Yihe; Foulsham, William et al. (2018) The immunoregulatory role of corneal epithelium-derived thrombospondin-1 in dry eye disease. Ocul Surf 16:470-477
Foulsham, William; Coco, Giulia; Amouzegar, Afsaneh et al. (2018) When Clarity Is Crucial: Regulating Ocular Surface Immunity. Trends Immunol 39:288-301
Hua, Jing; Inomata, Takenori; Chen, Yihe et al. (2018) Pathological conversion of regulatory T cells is associated with loss of allotolerance. Sci Rep 8:7059
Inomata, Takenori; Hua, Jing; Nakao, Takeshi et al. (2018) Corneal Tissue From Dry Eye Donors Leads to Enhanced Graft Rejection. Cornea 37:95-101
Chen, Yihe; Chauhan, Sunil K; Tan, Xuhua et al. (2017) Interleukin-7 and -15 maintain pathogenic memory Th17 cells in autoimmunity. J Autoimmun 77:96-103
Lee, Hyun Soo; Amouzegar, Afsaneh; Dana, Reza (2017) Kinetics of Corneal Antigen Presenting Cells in Experimental Dry Eye Disease. BMJ Open Ophthalmol 1:e000078
Inomata, Takenori; Mashaghi, Alireza; Hong, Jiaxu et al. (2017) Scaling and maintenance of corneal thickness during aging. PLoS One 12:e0185694
Chen, Yihe; Chauhan, Sunil K; Shao, Chunyi et al. (2017) IFN-?-Expressing Th17 Cells Are Required for Development of Severe Ocular Surface Autoimmunity. J Immunol 199:1163-1169
Dohlman, Thomas H; Ding, Julia; Dana, Reza et al. (2017) T Cell-Derived Granulocyte-Macrophage Colony-Stimulating Factor Contributes to Dry Eye Disease Pathogenesis by Promoting CD11b+ Myeloid Cell Maturation and Migration. Invest Ophthalmol Vis Sci 58:1330-1336
Mashaghi, Alireza; Marmalidou, Anna; Tehrani, Mohsen et al. (2016) Neuropeptide substance P and the immune response. Cell Mol Life Sci 73:4249-4264

Showing the most recent 10 out of 41 publications