The immune response requires a delicate balance between activation and attenuation. Likewise, therapies for autoimmune diseases and organ transplantation face the challenge of achieving enough immunosuppression to prevent organ rejection or limit autoreactivity without impairing the host's ability to protect against infections and malignancy. Regulatory T-cells (Treg) are an important T-cell subset crucial to self-tolerance, capable dampening or switching off antigen-specific immune responses (1). The transcription factor Forkheadbox-p3 (Foxp3) plays a key role in the development and functions of Treg. Foxp3 is regulated transcriptionally but also by post-translational modifications. We have shown that acetylation of lysine residues within the Foxp3 protein can enhance Treg function (2). Such acetylation also protects Foxp3 from proteasomal degradation and thus contributes to optimal Treg functions. Foxp3 acetylation is regulated by the competing actions of several histone/protein acetyltransferases (HAT) and histone/protein deacetylases (HDAC). We propose to study the role of Sirtuin-1 (Sirt1), a class III HDAC highly conserved across eukaryotic species and an important mediator of cellular metabolism and longevity, in Tregs. We have begun to exploit use of mice with targeted deletions of Sirt1, which is important as mice with global Sirt1 knockout suffer from metabolic problems and shortened lifespan. In addition, we have employed Sirt1 small molecule inhibitors to test the effect of transient Sirt1 inhibition in wild-type mice. Our preliminary data show that the targeted deletion of Sirt1 increases the acetylation and expression of Foxp3, and enhances the immunosuppressive functions of Treg. In addition, deletion of Sirt1 in Tregs, or its pharmacologic inhibition, attenuates allograft rejection and prolongs survival of murine cardiac allografts. Therefore, our central hypothesis is that targeting Sirt1 may have therapeutic value in autoimmunity and transplantation.
Our aims are to explore how Sirt1 deletion or inhibition: 1) improves allograft survival and function (in murine recipients with induced diabetes and renal failure);2) alleviates autoimmunity (in murine inflammatory bowel disease models);and, further, understand 3) distinct molecular mechanisms how Sirt1 influences T-cell biology beyond simply promoting Foxp3 acetylation. Our findings will likely prove important to the development of new immunomodulatory strategies for application in autoimmunity and transplantation. Our proposed studies are also important for an increased understanding of the functions of Sirt1 in immune responses, given the rising attention being given to the Sirt1 activator resveratrol (already available over the counter) and other more potent small molecule Sirt1 activators (3), for therapy of various diseases and for promotion of overall well-being. In addition, Sirt1 inhibitors are under consideration as anti-neoplastic drugs (4). Therefore, we caution that it is critical to learn more about the role of Sirt1 in the immune system as such treatment options are evolving.

Public Health Relevance

(provided by applicant): We study Sirtuin-1 as a novel therapeutic target to attenuate the immune response, which is relevant to improve treatment options to achieve immunosuppression, e.g. for patients suffering from self-reactive immune disease, or recipients of an organ transplant. Furthermore, it is important to learn more about the role of Sirtuin-1 in the immune system, as many investigators pursue Sirtuin-1 enhancement (diabetes mellitus, metabolic syndrome), or Sirtuin-1 suppression (certain cancers).

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Clinical Investigator Award (CIA) (K08)
Project #
1K08AI095353-01
Application #
8165446
Study Section
Allergy & Clinical Immunology-1 (AITC)
Program Officer
Prograis, Lawrence J
Project Start
2011-04-15
Project End
2016-03-31
Budget Start
2011-04-15
Budget End
2012-03-31
Support Year
1
Fiscal Year
2011
Total Cost
$129,951
Indirect Cost
Name
Children's Hospital of Philadelphia
Department
Type
DUNS #
073757627
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Wang, L; Beier, U H; Akimova, T et al. (2018) Histone/protein deacetylase inhibitor therapy for enhancement of Foxp3+ T-regulatory cell function posttransplantation. Am J Transplant 18:1596-1603
Lieber, Arnon D; Beier, Ulf H; Xiao, Haiyan et al. (2018) Loss of HDAC6 alters gut microbiota and worsens obesity. FASEB J :fj201701586R
Jiao, Jing; Han, Rongxiang; Hancock, Wayne W et al. (2017) Proximity Ligation Assay to Quantify Foxp3 Acetylation in Regulatory T Cells. Methods Mol Biol 1510:287-293
Huang, Jianbing; Wang, Liqing; Dahiya, Satinder et al. (2017) Histone/protein deacetylase 11 targeting promotes Foxp3+ Treg function. Sci Rep 7:8626
Laskin, Benjamin L; Jiao, Jing; Baluarte, H Jorge et al. (2017) The Effects of Tacrolimus on T-Cell Proliferation Are Short-Lived: A Pilot Analysis of Immune Function Testing. Transplant Direct 3:e199
Angelin, Alessia; Gil-de-Gómez, Luis; Dahiya, Satinder et al. (2017) Foxp3 Reprograms T Cell Metabolism to Function in Low-Glucose, High-Lactate Environments. Cell Metab 25:1282-1293.e7
Akimova, Tatiana; Levine, Matthew H; Beier, Ulf H et al. (2016) Standardization, Evaluation, and Area-Under-Curve Analysis of Human and Murine Treg Suppressive Function. Methods Mol Biol 1371:43-78
Xiao, Haiyan; Jiao, Jing; Wang, Liqing et al. (2016) HDAC5 controls the functions of Foxp3(+) T-regulatory and CD8(+) T cells. Int J Cancer 138:2477-86
Levine, Matthew H; Wang, Zhonglin; Xiao, Haiyan et al. (2016) Targeting Sirtuin-1 prolongs murine renal allograft survival and function. Kidney Int 89:1016-1026
Wang, Liqing; Kumar, Suresh; Dahiya, Satinder et al. (2016) Ubiquitin-specific Protease-7 Inhibition Impairs Tip60-dependent Foxp3+ T-regulatory Cell Function and Promotes Antitumor Immunity. EBioMedicine 13:99-112

Showing the most recent 10 out of 23 publications