T cells lacking HDAC11 have increased effector functions and mediate enhanced alloreactivity in a murine model

David M. Woods, Karrune V. Woan, Fengdong Cheng, Andressa L. Sodré, Dapeng Wang, Yongxia Wu, Zi Wang, Jie Chen, John Powers, Javier Pinilla-Ibarz, Yu Yu, Ya Zhang, Xuefeng Wu, Xiaoyan Zheng, Jeffrey Weber, Wayne W. Hancock, Edward Seto, Alejandro Villagra, Xue Zhong Yu, Eduardo M. Sotomayor

Research output: Contribution to journalArticlepeer-review

55 Scopus citations

Abstract

Histone acetylation and the families of enzymes responsible for controlling these epigenetic marks have been implicated in regulating T-cell maturation and phenotype. Here, we demonstrate a previously undefined role of histone deacetylase 11 (HDAC11) in regulating T-cell effector functions. Using EGFP-HDAC11 transgenic reporter mice, we found that HDAC11 expression was lower in effector relative to naive and central memory T-cell populations, and activation of resting T cells resulted in its decreased expression. Experiments using HDAC11 knockout (KO) mice revealed that T cells from these mice displayed enhanced proliferation, proinflammatory cytokine production, and effector molecule expression. In addition, HDAC11KO T cells had increased expression of Eomesodermin (Eomes) and TBX21 (Tbet), transcription factors previously shown to regulate inflammatory cytokine and effector molecule production. Conversely, overexpression of HDAC11 resulted in decreased expression of these genes. Chromatin immunoprecipitation showed the presence of HDAC11 at the Eomes and Tbet gene promoters in resting T cells, where it rapidly disassociated following T-cell activation. In vivo, HDAC11KO T cells were refractory to tolerance induction. HDAC11KO T cells also mediated accelerated onset of acute graft-versus-host disease (GVHD) in a murine model, characterized by increased proliferation of T cells and expression of interferon-γ, tumor necrosis factor, and EOMES. In addition, adoptive transfer of HDAC11KO T cells resulted in significantly reduced tumor burden in a murine B-cell lymphoma model. Taken together, these data demonstrate a previously unknown role of HDAC11 as a negative epigenetic regulator of T-cell effector phenotype and function.

Original languageEnglish (US)
Pages (from-to)146-155
Number of pages10
JournalBlood
Volume130
Issue number2
DOIs
StatePublished - Jul 13 2017

Bibliographical note

Funding Information:
The authors extend their appreciation to the Flow Cytometry Core, Tissue Core, and animal facilities at Moffitt Cancer Center for providing technical assistance. This work was supported by National Institutes of Health, National Cancer Institute, grants RO1CA179062 (E.M.S.), RO1CA134807 (E.M.S.), P50CA168536 (E.M.S.), R01CA143821 (X.-Z.Y.), and R01CA169116 (X.-Z.Y.).

Publisher Copyright:
© 2017 by The American Society of Hematology.

Fingerprint

Dive into the research topics of 'T cells lacking HDAC11 have increased effector functions and mediate enhanced alloreactivity in a murine model'. Together they form a unique fingerprint.

Cite this