The ability to distinguish self from non-self is essential in maintaining proper immune homeostasis. Most self-reactive T cells are eliminated in the thymus through negative selection;however some cells bearing T cell receptors that recognize self-proteins may still escape thymic selection and enter the peripheral system as mature T cells. To avoid autoimmunity, those cells must be inactivated. Anergy is of one of the mechanisms that ensure proper control of self-reactive T cells. CD4+ T cells become anergic or unresponsive to antigen following suboptimal or partial stimulation. In T helper cells, anergy is established as a consequence of the NFAT-dependent expression of a specific set of anergy-associated genes, which encode proteins that dampen TCR signaling and inhibit cytokine transcription. Several reports have also identified T cell anergy as an important mechanism of immune evasion induced by malignant tumors. Although evidence suggests that T cell anergy may play a key role in preventing autoimmune disease and facilitating the ability of cancer cells to evade immune responses, the precise role that T cell anergy plays in regulating T cell responses in vivo remains yet to be fully addressed. With this proposal we intend to further characterize the transcriptional mechanisms that regulate the induction of T cell anergy and use that knowledge to evaluate the specific role of T cell anergy in two different processes: maintenance of self tolerance and tumor-induced immune tolerance;and finally 3. Develop a strategy to regulate the induction of T cell anergy and evaluate its possible use a therapeutic tool. A detailed understanding of the regulation and the physiological role of T cell anergy and the identification of new targets to specifically modulate this process should provide valuable information to design strategies for the treatment of autoimmune diseases, to prevent graft rejection or to activate T cell responses against tumor cells or during chronic infections.

Public Health Relevance

Inactivation of self-reactive T cells has been identified as an important mechanism in the control of immune reactions against our own tissues. This process, termed anergy, is also crucial to understand how chronic infections and cancer cells can evade effective immune responses.
The aim of this project is to characterize the molecular mechanisms that control how T cells anergy is established and, based on that knowledge, develop potential therapeutic strategies that may help enhance T cell responses against cancer cells and chronic pathogens.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI059738-09
Application #
8436278
Study Section
Transplantation, Tolerance, and Tumor Immunology (TTT)
Program Officer
Lapham, Cheryl K
Project Start
2004-04-01
Project End
2015-03-31
Budget Start
2013-04-01
Budget End
2014-03-31
Support Year
9
Fiscal Year
2013
Total Cost
$386,199
Indirect Cost
$153,549
Name
Albert Einstein College of Medicine
Department
Pathology
Type
Schools of Medicine
DUNS #
110521739
City
Bronx
State
NY
Country
United States
Zip Code
10461
Mocholi, Enric; Dowling, Samuel D; Botbol, Yair et al. (2018) Autophagy Is a Tolerance-Avoidance Mechanism that Modulates TCR-Mediated Signaling and Cell Metabolism to Prevent Induction of T Cell Anergy. Cell Rep 24:1136-1150
Ames, Rachel Y; Ting, Li-Min; Gendlina, Inessa et al. (2017) The Transcription Factor NFAT1 Participates in the Induction of CD4+ T Cell Functional Exhaustion during Plasmodium yoelii Infection. Infect Immun 85:
Bandyopadhyay, Sanmay; Quinn, Thomas J; Scandiuzzi, Lisa et al. (2016) Low-Intensity Focused Ultrasound Induces Reversal of Tumor-Induced T Cell Tolerance and Prevents Immune Escape. J Immunol 196:1964-76
Cuervo, Ana Maria; Macian, Fernando (2014) Autophagy and the immune function in aging. Curr Opin Immunol 29:97-104
Shin, Daniel S; Jordan, Ayana; Basu, Samik et al. (2014) Regulatory T cells suppress CD4+ T cells through NFAT-dependent transcriptional mechanisms. EMBO Rep 15:991-9
Bandyopadhyay, Sanmay; Valdor, Rut; Macian, Fernando (2014) Tle4 regulates epigenetic silencing of gamma interferon expression during effector T helper cell tolerance. Mol Cell Biol 34:233-45
Baine, Ian; Basu, Samik; Ames, Rachel et al. (2013) Helios induces epigenetic silencing of IL2 gene expression in regulatory T cells. J Immunol 190:1008-16
Abe, Brian T; Macian, Fernando (2013) Uncovering the mechanisms that regulate tumor-induced T-cell anergy. Oncoimmunology 2:e22679
Tsiperson, Vladislav; Gruber, Ross C; Goldberg, Michael F et al. (2013) Suppression of inflammatory responses during myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis is regulated by AKT3 signaling. J Immunol 190:1528-39
Valdor, Rut; Macian, Fernando (2013) Induction and stability of the anergic phenotype in T cells. Semin Immunol 25:313-20

Showing the most recent 10 out of 24 publications