IL-35 is a newly discovered cytokine that contributes to the suppressive capacity of Tregs. IL-35 Is a heterodimer consisting of an alpha (p35) and beta chain (Ebl3). It is a member of the IL-12 cytokine family, which is characterized by the sharing of 3 alpha (pi 9, p28, p35) and 2 beta (p40 and Ebi3) subunits, and also includes IL-12 (p35/p40), IL-23 (p19/p40) and IL-27 (p28/Ebi3). Additional information regarding the basis of subunit sharing, dimer formation, and other mechanisms to regulate IL-12 family members at the protein level will be critical for the development of IL-35-based therapeutics that do not interfere with function of IL-12 or IL-27. The objective of this research plan is to characterize the Interactions between IL-12 family subunits, particulariy IL-35 and the subunits that it shares with IL-12 and IL-27. This work will address the following questions: Alm#1: Is IL-35 secretion regulated at the protein level? My preliminary data indicate that IL-35 is poorly secreted by transfected cells, compared to IL-12 and IL-27, suggesting IL-35 formation is unfavorable. We will determine if activated dendritic cells, which express IL-12 and IL-27 are capable of secreting IL-35 by, IP/western blot. We will evaluate the ability of multiple cell types to ectopically express IL- 35, to determine if an unknown Treg-specific factor is required for optimal IL-35 production. We will assess relative chain pairing efficiencies within the IL-12 family by comparing intracellular and secreted cytokine levels in a normalized in vitro expression system by IP/western blot. Finally, we will monitor intracellular competition between IL-12 family subunits with a shared binding partner in FRET and BiFC assays.
Aim#2 : Do IL-35 subunits use the same binding interface for formation of IL-12 and IL-27. The development of theraputic applications of IL-35 that do not interfere with IL-12 and IL-27 will require extensive knowledge of the shared dimerization interfaces of each subunit. We will mutate p35 residues that are involved in formation of the IL-12 interface and assess their role in IL-35 formation using IP/western blot and functional assays. We will also mutate the putative binding interface of Ebi3 and determine if the same residues are involved in dimerization of both IL-27 and IL-35. Finally, we will attempt to identify residues in both p35 and Ebl3 that can abbrogate IL-35 formation and leave IL-12 and IL-27 function intact. Regulatory T cells play an important role in suppression of autoimmune diseases, and lack of Treg function can promote anti-tumor Immunity. To utilize Tregs to control disease, it is important to understand proteins that mediate their function. We will investigate the mechanisms that regulate expression of Treg- speclfic cytokine IL-35, a potential theraputic target for autoimmune diseases and cancer.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Postdoctoral Individual National Research Service Award (F32)
Project #
1F32AI084330-01
Application #
7749788
Study Section
Special Emphasis Panel (ZRG1-F07-E (20))
Program Officer
Prograis, Lawrence J
Project Start
2009-09-16
Project End
2012-09-15
Budget Start
2009-09-16
Budget End
2010-09-15
Support Year
1
Fiscal Year
2009
Total Cost
$45,218
Indirect Cost
Name
St. Jude Children's Research Hospital
Department
Type
DUNS #
067717892
City
Memphis
State
TN
Country
United States
Zip Code
38105
Jones, Lindsay L; Chaturvedi, Vandana; Uyttenhove, Catherine et al. (2012) Distinct subunit pairing criteria within the heterodimeric IL-12 cytokine family. Mol Immunol 51:234-44
Jones, Lindsay L; Vignali, Dario A A (2011) Molecular interactions within the IL-6/IL-12 cytokine/receptor superfamily. Immunol Res 51:5-14