Abstract

T-regulatory cells are an upsurge in the tumor microenvironment and induce immune-evasion. CD4+ Treg cells are well characterized whereas the role of CD8+ Tregs in cancer has recently started to crease attention. Here, we report an augmentation CD8+FOXP3+ Tregs in breast tumor microenvironment. FOXP3, the lineage-specific transcription factor, is a dominant regulator of Treg cell development and function. FOXP3 is induced preferentially by divergent signaling in CD4+ Treg cells. But how FOXP3 is induced and maintained in tumor-CD8+ Tregs is the Cinderella of the investigation. We observed that RUNX3, a CD8+ lineage-specific transcription factor, binds at the FOXP3-promoter to induce its transcription. In addition to promoter activation, involvement of cis-elements CNS1 and CNS2 in the transcriptional regulation of FOXP3 was also evident in these cells. SMAD3 binds to CNS1 region and acts as transcription inducer, whereas GATA3 plays a temporal role in the FOXP3 transcription by differential chromatin modification in CNS regions. In CNS1 region, GATA3 acts as a repressor for FOXP3 in naïve CD8+ T cells. Whereas in CD8+ Tregs, GATA3 binds directly at CNS2 region and persuaded the maintenance of FOXP3. Therefore, the intervention of these concerted transcriptional machinery may have a therapeutic potential in immunotherapy of cancer.

Highlights

  • T-regulatory cells (Tregs) play a pivotal role in the development and maintenance of peripheral immune tolerance as defects in their compartment lead to severe autoimmune diseases

  • We disclosed the mechanisms of development and function of CD8+ Treg cells in breast tumor microenvironment, emphasizing the transcriptional regulation of their FOXP3 gene

  • Most of the previous studies have described the regulation of FOXP3 in CD4+ Treg cells; CD4+ and CD8+ cells have different lineage-specific transcription machinery which suggests that the regulation of FOXP3 in CD4+ and CD8+ Treg cells can differ

Read more

Summary

Introduction

T-regulatory cells (Tregs) play a pivotal role in the development and maintenance of peripheral immune tolerance as defects in their compartment lead to severe autoimmune diseases. In CD4+ Treg cells, FOXP3 induction is coupled with the activation of TCR/NFAT-NFκB-signalling or by TGFβ/SMAD3-signaling[2, 10] In addition to these factors other, transcription factors, viz; GATA3, RUNX3, and RUNX1 play pivotal roles in the regulation of Foxp[3] gene expression[11, 12]. Since CTLA4 is transcriptionally activated by FOXP3, we used this co-stimulatory molecule as a surface signature for the isolation of tumor-CD8+ Treg cells for our study Exploiting these cells we could successfully show that transcriptional activation of FOXP3-promoter is associated with chromatin modification and binding of SMAD3, GATA3, and RUNX3 at the different non-coding conserved sequence (CNS) and the promoter regions of FOXP3. This study may open a new way to target CD8+ Treg cells and potentiates the antitumor immunity during cancer immunotherapy

Methods
Results
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.