MiR-146b antagomir-treated human Tregs acquire increased GVHD inhibitory potency

Yunjie Lu, Keli L Hippen, Amanda L. Lemire, Jian Gu, Weizhi Wang, Xuhao Ni, Parvathi Ranganathan, Bruce L. Levine, James L. Riley, Carl H. June, Laurence A. Turka, David H. Munn, Ramiro Garzon, Ling Lu, Bruce R Blazar

Research output: Contribution to journalArticlepeer-review

51 Scopus citations

Abstract

CD4+CD25+FoxP3+ thymic-derived regulatory T cells (tTregs) are indispensable for maintaining immune system equilibrium. Adoptive transfer of tTregs is an effective means of suppressing graft-versus-host disease (GVHD) in murine models and in early human clinical trials. Tumor necrosis factor receptor-associated factor 6 (TRAF6), an ubiquitin-conjugating enzyme that mediates nuclear factor κB (NF-κB) activation, plays an essential role in modulating regulatory T cell survival and function. MicroRNAs (miRNAs) are noncoding RNAs, which mediate RNA silencing and posttranscriptional gene repression. By performing comprehensive TaqMan Low Density Array miRNA assays, we identified 10 miRNAs differentially regulated in human tTreg compared with control T cells. One candidate, miR-146b, is preferentially and highly expressed in human naive tTregs compared with naive CD4 T cells. miRNA prediction software revealed that TRAF6 was the one of the top 10 scored mRNAs involved tTreg function with the highest probability as a potential miR-146b target. Antagomir-mediated knockdown of miRNA- 146b, but not another miRNA-146 family member (miRNA-146a), enhanced TRAF6 expression. TRAF6, in turn, increases NF-κB activation, which is essential for tTreg function as well as Foxp3 protein and antiapoptotic gene expression, and downregulates proapoptotic gene expression. miR-146b knockdown increased the nuclear localization and expression of genes regulated by NF-κB, which was associated with enhanced tTreg survival, proliferation, and suppressive function measured in vitro and in vivo. TRAF6 inhibition had the opposite effects. We conclude that an miR-146b-TRAF6-NF-κB-FoxP3 signaling pathway restrains regulatory T cell survival, proliferation, and suppressor function. In vitro exposure of human tTregs to miR-146b antagomirs can be exploited to improve the clinical efficacy of human adoptive tTreg transfer in a GVHD setting.

Original languageEnglish (US)
Pages (from-to)1424-1435
Number of pages12
JournalBlood
Volume128
Issue number10
DOIs
StatePublished - Sep 8 2016

Bibliographical note

Funding Information:
This work was supported in part by research grants from the Children's Cancer Research Fund (K.L.H. and B.R.B), Leukemia and Lymphoma Translational Research Grant R6029-07 (B.R.B.), National Institutes of Health (NIH) National Heart, Lung, and Blood Institute grant R01 HL11879 (B.R.B.), NIH National Cancer Institute (NCI) grant P01 CA067493 (B.R.B.), an NIH Clinical and Translational Science Award to the University of Minnesota (8UL1TR000114), and NIH/NCI grant P30 CA77598 utilizing the shared resource Flow Cytometry Core from the Masonic Cancer Center, University of Minnesota. This work was also supported in part by National Natural Science Fund grant 81571564 (L.L.), National Natural Science Fund: Outstanding Youth Fund grant 81522020 (L.L.), 863 Young Scientists Special Fund grant SS2015AA020932 (L.L.), and Major Program of the National Natural Science Foundation of China grant 91442117 (L.L.).

Publisher Copyright:
© 2016 by The American Society of Hematology.

Fingerprint

Dive into the research topics of 'MiR-146b antagomir-treated human Tregs acquire increased GVHD inhibitory potency'. Together they form a unique fingerprint.

Cite this