Abstract

Simple SummaryThe gastrointestinal tumor microenvironment is regulated by cytokine production from the tumor and adjacent immune cells. G-CSFR is a cytokine that is highly produced in the gastrointestinal tumor microenvironment and may have unrecognized pro-tumorigenic activities. Here, we explored the impact of G-CSF and its receptor on macrophage responses in colon and pancreas tumors in mice. We found G-CSF/G-CSFR to promote a pro-tumorigenic macrophage response. Upon deletion of G-CSFR, macrophages exhibited increased tumor killing ability in culture and decreased tumor growth in mice. Our findings suggest that G-CSFR blockade may be beneficial to promoting anti-tumorigenic macrophage responses and should be further examined as a therapeutic target.Tumor-associated macrophages (TAMs) in the gastrointestinal tumor microenvironment (TME) are known to polarize into populations exhibiting pro- or anti-tumoral activity in response to stimuli such as growth factors and cytokines. Our previous work has recognized granulocyte colony-stimulating factor (G-CSF) as a cytokine capable of influencing immune cells of the TME exhibiting pro-tumoral activity. Here, we aimed to focus on how G-CSF regulates TAM phenotype and function and the effects on gastrointestinal (GI) tumor progression. Thus, wildtype (WT) and G-CSFR−/− macrophages were examined for cytokine production, gene expression, and transcription factor activity. Adoptive transfer of WT or G-CSFR−/− macrophages into tumor-bearing mice was performed to study their influence in the progression of colon (MC38) and pancreatic (PK5L1940) tumor mouse models. Finally, the difference in cytotoxic potential between WT and G-CSFR−/− macrophages was examined both in vitro and in vivo. Our results indicate that G-CSF promotes increased IL-10 production and decreased IL-12 production, which was reversed in G-CSFR−/− macrophages for a pro-inflammatory phenotype. Furthermore, G-CSFR−/− macrophages were characterized by higher levels of NOS2 expression and NO production, which led to greater tumor related cytotoxicity both in vitro and in vivo. Our results suggest that in the absence of G-CSFR, macrophage-related tumor cytotoxicity was amplified. These findings, along with our previous reports, pinpoint G-CSF /G-CSFR as a prominent target for possible clinical applications that aim to control the TME and the GI tumor progression.

Highlights

  • Gastrointestinal cancers, and pancreatic and colorectal cancer (CRC), are aggressive malignant diseases with a low survival rate and poor prognosis for late stage disease.Colon cancer is the third most common cancer worldwide with grave impacts on global health

  • For WT BMM, we found that the CD115- population was G-CSFR+, and upon activation with LPS, CD115 expression was lost (Figure S1B)

  • We showed that two more cytokines, IL-1β and TNF-α were detected in high levels in G-CSFR−/− BMMs, while in activated WT BMMs treated with exogenous granulocyte colony-stimulating factor (G-CSF), a dramatically decreased production of those cytokines was observed

Read more

Summary

Introduction

Gastrointestinal cancers, and pancreatic and colorectal cancer (CRC), are aggressive malignant diseases with a low survival rate and poor prognosis for late stage disease.Colon cancer is the third most common cancer worldwide with grave impacts on global health. Gastrointestinal cancers, and pancreatic and colorectal cancer (CRC), are aggressive malignant diseases with a low survival rate and poor prognosis for late stage disease. Causes of GI cancer have been linked to environmental, nutritional, and genetic/epigenetic factors, and to age and chronic inflammation diseases [1]. Within GI tumors, chronic inflammatory conditions are usually present with several immune cells having been identified in the tumor microenvironment (TME), including T cells (both cytotoxic and helper cells), mesenchymal stem cells, and dendritic cells, as well as tumor-associated macrophages (TAMs) [2,3]. TAMs are highly infiltrative and have been identified in high numbers in the CRC and pancreatic cancer TME [5,6]. M1 macrophages, described as classically activated, are polarized by microenvironment signals such as interferon-gamma (IFN-γ) promoting signal transducer and activator of transcription 1 (STAT1) signaling and lipopolysaccharide (LPS)

Objectives
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.