Immunological memory is a cardinal property of the adaptive immune system and memory CD8 T cells play a critical role in providing long-term protective immunity against reoccurring infectious disease and cancers. Generating long-lived functional memory T cells is vital for any successful vaccination campaign. For the greatest protection possible memory T cells must possess 1) longevity, 2) high proliferative potential and 3) rapid development of effector functions when antigen is reencountered. It is not known how these properties are instilled into the precursors of memory T cells during immune responses, and this is the primary focus of this proposal. Our prior studies using acute viral and bacterial infection in mice embarked on identification of the memory CD8 T cell precursors, and this work helped to distinguish this subset of effector cells from those that will die. Our recent preliminary results outline formation of memory precursor effector cell (MPEC) and short-lived effector cell (SLEC) lineages during infection and delineate when effector CD8 T cells begin to adopt the SLEC fate. We found that the transcription factor T-bet is a key lineage-determining factor that controls the SLEC/MPEC fate decision during infection, and that the relative concentration of T-bet in the effector CD8 T cells modulated this process. High levels of T-bet specified SLEC development whereas low levels specified MPEC development. Furthermore, we found that IL-12 was a potent inducer of T-bet expression and SLECs during infection. Thus, early inflammatory signals not only define the types of effector CD8 T cells that form and enhance their functions during infection, but they also influence their longevity and ability to develop into memory CD8 T cells that can self-renew. This highlights a novel control mechanism by which the innate immune system regulates T cell homeostasis. The over-arching goal of this proposal is to elucidate the mechanisms that determine which effector CD8 T cells become MPECs or SLECs during immune responses in order to create new avenues for improving vaccination and immunotherapies that fight infectious disease and cancer. Specifically, we aim to (i) determine when T-bet functions during effector CD8 T cell differentiation and how T-bet and IL-12 signals are coordinately integrated, (ii) identify other cytokines that regulate T-bet expression and terminal effector cell differentiation, (iii) determine if T-bet controls the memory cell potential of Th1 effector CD4 T cells during LCMV infection.

Public Health Relevance

As acute infections resolve, effector CD8 T cells differentiate into short-lived effector cells (SLECs) and memory precursor effector cells (MPECs) capable of generating long-lived, protective memory CD8 T cells.
We aim to identify the genetic pathways that control this process and have found the transcription factor T- bet is an important modulator of memory CD8 T cell development. Here, we propose to delineate when and how T-bet functions in activated CD8 T cells, to identify the inflammatory signals that control T-bet expression during infection with Biodefense Priority Pathogens, and to determine if T-bet plays a similar role in memory CD4 T cell development.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI074699-02
Application #
7879509
Study Section
Cellular and Molecular Immunology - B Study Section (CMIB)
Program Officer
Lapham, Cheryl K
Project Start
2009-06-25
Project End
2014-05-31
Budget Start
2010-06-01
Budget End
2011-05-31
Support Year
2
Fiscal Year
2010
Total Cost
$383,304
Indirect Cost
Name
Yale University
Department
Microbiology/Immun/Virology
Type
Schools of Medicine
DUNS #
043207562
City
New Haven
State
CT
Country
United States
Zip Code
06520
Guan, Tianxia; Dominguez, Claudia X; Amezquita, Robert A et al. (2018) ZEB1, ZEB2, and the miR-200 family form a counterregulatory network to regulate CD8+ T cell fates. J Exp Med 215:1153-1168
Ho, Ping-Chih; Kaech, Susan M (2017) Reenergizing T cell anti-tumor immunity by harnessing immunometabolic checkpoints and machineries. Curr Opin Immunol 46:38-44
Xin, Annie; Masson, Frederick; Liao, Yang et al. (2016) A molecular threshold for effector CD8(+) T cell differentiation controlled by transcription factors Blimp-1 and T-bet. Nat Immunol 17:422-32
Laidlaw, Brian J; Craft, Joseph E; Kaech, Susan M (2016) The multifaceted role of CD4(+) T cells in CD8(+) T cell memory. Nat Rev Immunol 16:102-11
Chen, Jonathan H; Perry, Curtis J; Tsui, Yao-Chen et al. (2015) Prostaglandin E2 and programmed cell death 1 signaling coordinately impair CTL function and survival during chronic viral infection. Nat Med 21:327-34
Marshall, Heather D; Ray, John P; Laidlaw, Brian J et al. (2015) The transforming growth factor beta signaling pathway is critical for the formation of CD4 T follicular helper cells and isotype-switched antibody responses in the lung mucosa. Elife 4:e04851
Dominguez, Claudia X; Amezquita, Robert A; Guan, Tianxia et al. (2015) The transcription factors ZEB2 and T-bet cooperate to program cytotoxic T cell terminal differentiation in response to LCMV viral infection. J Exp Med 212:2041-56
Laidlaw, Brian J; Cui, Weiguo; Amezquita, Robert A et al. (2015) Production of IL-10 by CD4(+) regulatory T cells during the resolution of infection promotes the maturation of memory CD8(+) T cells. Nat Immunol 16:871-9
Ho, Ping-Chih; Bihuniak, Jessica Dauz; Macintyre, Andrew N et al. (2015) Phosphoenolpyruvate Is a Metabolic Checkpoint of Anti-tumor T Cell Responses. Cell 162:1217-28
Gray, Simon M; Kaech, Susan M; Staron, Matthew M (2014) The interface between transcriptional and epigenetic control of effector and memory CD8? T-cell differentiation. Immunol Rev 261:157-68

Showing the most recent 10 out of 26 publications