Dry eye disease (DED), a chronic immune-mediated disorder of the ocular surface, is characterized by sustained ocular surface inflammation and disruption of the ocular surface epithelium, which in severe cases can lead to corneal scarring and blindness. DED has a very high prevalence, almost triple that of age-related macular degeneration, affecting many millions of individuals in the United States alone, and its prevalence is set to increase considerably with the aging of the population. Current expenditures for treating DED surpass $2 billion dollars annually, and because it often affects visual function in working adults, it leads to lost productivity by impacting job performance. While the role of immunity in amplifying DED severity has been known for some time, it is only recently that we have gained insights into the immunopathogenic mechanisms that drive DED. Evidence from several laboratories, including the Principal Investigator's, has established strong evidence for the critical role of T helper-17 (Th17) cells, in driving immune-mediated damage to the ocular surface in DED. More recent evidence from the PI's lab shows that DED can be maintained chronically for months, even after termination of desiccating stress to the eye, by memory Th17 cells, which are distinct from the effector T cells that mediate disease after acute ocular desiccating stress. In addition, data from the PI's lab demonstrate that in DED the regulatory T cells (Tregs), which are normally highly effective in suppressing T cell-mediated inflammation, are particularly defective in suppressing the Th17 response. While these data provide new insights into DED immunopathogenesis, numerous important questions remain unanswered. We hypothesize that DED is characterized by a microenvironment that promotes the generation and maintenance of Th17 immunity, which in turn leads to suppressed function of Tregs that normally maintain immune quiescence. The principal objectives of this project are to (i) define the factors that influence the generation and maintenance of immunologic memory, and characterize precisely the dysfunction of (ii) the corneal epithelium and (iii) Tregs in maintaining immune homeostasis in DED. To achieve these major objectives, three specific aims have been defined to answer the following questions:
Aim 1 : What are the critical cytokine mechanisms that lead to induction and maintenance of Th17 immunological memory in chronic dry eye disease? Aim 2: How does epithelial damage affect the immunomodulatory function of the corneal epithelium in DED? And, Aim 3: Which among the Th17- associated cytokines interact directly with Tregs to suppress their regulatory function? 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 precise immunopathogenesis, and the relative dearth of effective treatments.

Public Health Relevance

Dry eye disease is an extremely common inflammatory disorder of the ocular surface associated with visual dysfunction, with as of yet incompletely understood immunopathogenic mechanisms. We propose to determine the specific immune-based cellular and molecular pathogenic elements that amplify dry eye disease severity and promote chronic immune based inflammation. The hope is that better understanding of these pathogenic mechanisms will yield important therapeutic targets that could be used in better management of the many millions of Americans suffering from dry eye disease.

Agency
National Institute of Health (NIH)
Institute
National Eye Institute (NEI)
Type
Research Project (R01)
Project #
5R01EY020889-06
Application #
8928621
Study Section
Diseases and Pathophysiology of the Visual System Study Section (DPVS)
Program Officer
Mckie, George Ann
Project Start
2010-09-30
Project End
2019-08-31
Budget Start
2015-09-01
Budget End
2016-08-31
Support Year
6
Fiscal Year
2015
Total Cost
Indirect Cost
Name
Schepens Eye Research Institute
Department
Type
DUNS #
073826000
City
Boston
State
MA
Country
United States
Zip Code
Inomata, Takenori; Hua, Jing; Nakao, Takeshi et al. (2018) Corneal Tissue From Dry Eye Donors Leads to Enhanced Graft Rejection. Cornea 37:95-101
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
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