Abstract

The phagocyte NADPH oxidase generates superoxide anion and downstream reactive oxidant intermediates in response to infectious threat, and is a critical mediator of antimicrobial host defense and inflammatory responses. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that are recruited by cancer cells, accumulate locally and systemically in advanced cancer, and can abrogate anti-tumor immunity. Prior studies have implicated the phagocyte NADPH oxidase as being an important component promoting MDSC accumulation and immunosuppression in cancer. We therefore used engineered NADPH oxidase-deficient (p47phox−/−) mice to delineate the role of this enzyme complex in MDSC accumulation and function in a syngeneic mouse model of epithelial ovarian cancer. We found that the presence of NADPH oxidase did not affect tumor progression. The accumulation of MDSCs locally and systemically was similar in tumor-bearing wild-type (WT) and p47phox−/− mice. Although MDSCs from tumor-bearing WT mice had functional NADPH oxidase, the suppressive effect of MDSCs on ex vivo stimulated T cell proliferation was NADPH oxidase-independent. In contrast to other tumor-bearing mouse models, our results show that MDSC accumulation and immunosuppression in syngeneic epithelial ovarian cancer is NADPH oxidase-independent. We speculate that factors inherent to the tumor, tumor microenvironment, or both determine the specific requirement for NADPH oxidase in MDSC accumulation and function.

Highlights

  • Inflammatory cells that constitute the cancer microenvironment can limit or stimulate tumor growth

  • NADPH oxidase does not affect overall survival in mice with ovarian cancer To evaluate the role of NADPH oxidase in regulating ovarian tumor growth, we challenged WT and NADPH oxidase-deficient p47phox2/2 mice with intraperitoneal Mouse ovarian surface epithelial cancer (MOSEC)

  • Because phagocyte oxidase proteins could have NADPH oxidase-independent signaling, we evaluated survival following tumor challenge in another NADPH oxidase-deficient model [31], and found no significant difference compared to WT mice

Read more

Summary

Introduction

Inflammatory cells that constitute the cancer microenvironment can limit or stimulate tumor growth. MDSCs can suppress anti-tumor responses through several mechanisms: suppression of CD4+ and CD8+ T cells by arginine and cysteine depletion, inhibition of T cell recruitment to tumor sites, inhibition of T cell-peptide-MHC interactions, skewing of the cytokine milieu toward type 2 or regulatory responses, and modulating NK and NKT responses [3,4,5,6,7,8,9,10,11,12,13] In addition to their immunosuppressive properties, MDSCs can secrete factors (e.g., vascular endothelial growth factor (VEGF)) that enhance tumor growth, invasion, and metastasis [5,14]

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.