Abstract

BackgroundCD4+ T cells play multiple roles in controlling tumor growth and increasing IFN-γ+ T-helper 1 cell population could promote cell-mediated anti-tumor immune response. We have previously showed that low-dose DNA demethylating agent decitabine therapy promotes CD3+ T-cell proliferation and cytotoxicity; however, direct regulation of purified CD4+ T cells and the underlying mechanisms remain unclear.MethodsThe effects of low-dose decitabine on sorted CD4+ T cells were detected both in vitro and in vivo. The activation, proliferation, intracellular cytokine production and cytolysis activity of CD4+ T cells were analyzed by FACS and DELFIA time-resolved fluorescence assays. In vivo ubiquitination assay was performed to assess protein degradation. Moreover, phosphor-p65 and IκBα levels were detected in sorted CD4+ T cells from solid tumor patients with decitabine-based therapy.ResultsLow-dose decitabine treatment promoted the proliferation and activation of sorted CD4+ T cells, with increased frequency of IFN-γ+ Th1 subset and enhanced cytolytic activity in vitro and in vivo. NF-κB inhibitor, BAY 11-7082, suppressed decitabine-induced CD4+ T cell proliferation and IFN-γ production. In terms of mechanism, low-dose decitabine augmented the expression of E3 ligase β-TrCP, promoted the ubiquitination and degradation of IκBα and resulted in NF-κB activation. Notably, we observed that in vitro low-dose decitabine treatment induced NF-κB activation in CD4+ T cells from patients with a response to decitabine-primed chemotherapy rather than those without a response.ConclusionThese data suggest that low-dose decitabine potentiates CD4+ T cell anti-tumor immunity through enhancing IκBα degradation and therefore NF-κB activation and IFN-γ production.

Highlights

  • Immunotherapy has become a standard approach for the treatment of some types of cancers and has the potential to control tumor development

  • We observed that in vitro low-dose decitabine treatment induced NF-κB activation in CD4+ T cells from patients with a response to decitabine-primed chemotherapy rather than those without a response. These data suggest that low-dose decitabine potentiates CD4+ T cell antitumor immunity through enhancing IκBα degradation and NF-κB activation and IFN-γ production

  • Based on their functions and cytokine-producing patterns, CD4+ T helper cells are comprised of different functional subsets, including IFN-γ-producing T-helper 1 (Th1), T-helper 2 (Th2), T-helper 17 (Th17) and regulatory T cells (Tregs), which carry out specialized immunoregulatory functions to either enhance or inhibit immune response (Kim and Cantor, 2014)

Read more

Summary

Introduction

Immunotherapy has become a standard approach for the treatment of some types of cancers and has the potential to control tumor development. CD4+ T helper cells are recognized to be required for the formation of CD8+ CTLs. Currently, numerous studies have demonstrated that CD4+ T cells actively participate in shaping anti-tumor immunity (Kim and Cantor, 2014; Zander et al, 2019). Numerous studies have demonstrated that CD4+ T cells actively participate in shaping anti-tumor immunity (Kim and Cantor, 2014; Zander et al, 2019) Based on their functions and cytokine-producing patterns, CD4+ T helper cells are comprised of different functional subsets, including IFN-γ-producing T-helper 1 (Th1), T-helper 2 (Th2), T-helper 17 (Th17) and regulatory T cells (Tregs), which carry out specialized immunoregulatory functions to either enhance or inhibit immune response (Kim and Cantor, 2014). We have previously showed that low-dose DNA demethylating agent decitabine therapy promotes CD3+ T-cell proliferation and cytotoxicity; direct regulation of purified CD4+ T cells and the underlying mechanisms remain unclear

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.