Despite extraordinary medical advances in the last century, infectious diseases remain the second leading cause of death worldwide, and disease-associated morbidity causes immeasurable harm. The development of new vaccines is, therefore, an imperative for improving global health. The ability of adaptive immune cells to respond more rigorously after a primary exposure, preventing pathology, and in essence ?remembering? the infection is the basis for vaccination. A heterogeneous population of long-lived memory T cells mediate protection from reinfection with previously encountered pathogens; keep chronic, opportunistic and latent pathogens at bay; and defend against tumor growth and metastases. Thus, understanding how memory lymphocytes are induced and sustained is of central importance in the rational design of new vaccines. Tissue- resident memory T cells provide sentinel protection at body surfaces such as the intestinal epithelium, and provide a first line of adaptive immune defense to reinfection. While we now know that tissue-resident T memory cells provide an essential component of immune memory, the transcriptional pathways regulating their formation, survival, and function, and how these relate to those that promote the formation of other memory populations are poorly understood. Improving our understanding of these topics will allow us to harness the immediate protective capacity of this vital memory T cell population in tissues where infections typically begin and modulate activity in the context of immunopathology. To this end, we present a preliminary in vivo functional screen that identifies numerous new regulators of CD8+ tissue-resident memory T cell differentiation. Based on these novel findings, we propose to: (1) Study how Runx3 and Blimp1 support the formation, homeostasis, and function of the CD8+ tissue-resident memory T cell population. (2) Delineate the role of the bromodomain and extraterminal domain (BET) epigenetic ?reader? of histone acetylation, Brd4, in regulating gene expression during memory T cell differentiation. (3) Define the transcriptional network that regulates CD8+ tissue-resident memory T cell formation. By using single-cell sequencing and comparison of open chromatin regions in CD8+ tissue-resident memory T cell populations to predict important transcriptional regulators, we will expand our in vivo screening strategy to identify regulators of CD8+ tissue-resident memory cells. We will leverage the results to enhance tissue-resident memory activity in the context of tumor growth and viral infection. By discovering the transcriptional program and molecular regulators promoting differentiation, survival, and function of tissue- resident memory T cells, we will identify novel targets that can be exploited in the strategic design of therapeutic and protective vaccines, the development of which are of crucial importance to human health.

Public Health Relevance

Understanding how long-lived protective immunity mediated by CD8+ T cells is established and sustained in tissues is of significant importance to human health. By identifying the unique transcriptional pathways and regulators of tissue-resident memory CD8+ T cells, a newly appreciated and critical memory T cell population, we will discover novel targets that can be exploited to improve the design of therapeutic and protective vaccines.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI067545-13
Application #
9731374
Study Section
Cellular and Molecular Immunology - A Study Section (CMIA)
Program Officer
Kelly, Halonna R
Project Start
2006-06-01
Project End
2022-06-30
Budget Start
2019-07-01
Budget End
2020-06-30
Support Year
13
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of California, San Diego
Department
Biology
Type
Schools of Arts and Sciences
DUNS #
804355790
City
La Jolla
State
CA
Country
United States
Zip Code
92093
Omilusik, Kyla D; Nadjsombati, Marija S; Shaw, Laura A et al. (2018) Sustained Id2 regulation of E proteins is required for terminal differentiation of effector CD8+ T cells. J Exp Med 215:773-783
Delpoux, Arnaud; Michelini, Rodrigo Hess; Verma, Shilpi et al. (2018) Continuous activity of Foxo1 is required to prevent anergy and maintain the memory state of CD8+ T cells. J Exp Med 215:575-594
Widjaja, Christella E; Olvera, Jocelyn G; Metz, Patrick J et al. (2017) Proteasome activity regulates CD8+ T lymphocyte metabolism and fate specification. J Clin Invest 127:3609-3623
Phan, Anthony T; Goldrath, Ananda W; Glass, Christopher K (2017) Metabolic and Epigenetic Coordination of T Cell and Macrophage Immunity. Immunity 46:714-729
Yu, Bingfei; Zhang, Kai; Milner, J Justin et al. (2017) Epigenetic landscapes reveal transcription factors that regulate CD8+ T cell differentiation. Nat Immunol 18:573-582
Stradner, Martin H; Cheung, Kitty P; Lasorella, Anna et al. (2016) Id2 regulates hyporesponsive invariant natural killer T cells. Immunol Cell Biol 94:640-5
Shaw, Laura A; BĂ©langer, Simon; Omilusik, Kyla D et al. (2016) Id2 reinforces TH1 differentiation and inhibits E2A to repress TFH differentiation. Nat Immunol 17:834-43
Chea, Sylvestre; Schmutz, Sandrine; Berthault, Claire et al. (2016) Single-Cell Gene Expression Analyses Reveal Heterogeneous Responsiveness of Fetal Innate Lymphoid Progenitors to Notch Signaling. Cell Rep 14:1500-1516
Almaden, Jonathan V; Liu, Yi C; Yang, Edward et al. (2016) B-cell survival and development controlled by the coordination of NF-?B family members RelB and cRel. Blood 127:1276-86
Stone, Erica L; Pepper, Marion; Katayama, Carol D et al. (2015) ICOS coreceptor signaling inactivates the transcription factor FOXO1 to promote Tfh cell differentiation. Immunity 42:239-251

Showing the most recent 10 out of 36 publications