Abstract

In the tumor microenvironment, local immune dysregulation is driven in part by macrophages and dendritic cells that are polarized to a mixed proinflammatory/immune-suppressive phenotype. The unfolded protein response (UPR) is emerging as the possible origin of these events. Here we report that the inositol-requiring enzyme 1 (IRE1α) branch of the UPR is directly involved in the polarization of macrophages in vitro and in vivo, including the up-regulation of interleukin 6 (IL-6), IL-23, Arginase1, as well as surface expression of CD86 and programmed death ligand 1 (PD-L1). Macrophages in which the IRE1α/X-box binding protein 1 (Xbp1) axis is blocked pharmacologically or deleted genetically have significantly reduced polarization and CD86 and PD-L1 expression, which was induced independent of IFNγ signaling, suggesting a novel mechanism in PD-L1 regulation in macrophages. Mice with IRE1α- but not Xbp1-deficient macrophages showed greater survival than controls when implanted with B16.F10 melanoma cells. Remarkably, we found a significant association between the IRE1α gene signature and CD274 gene expression in tumor-infiltrating macrophages in humans. RNA sequencing (RNASeq) analysis showed that bone marrow-derived macrophages with IRE1α deletion lose the integrity of the gene connectivity characteristic of regulated IRE1α-dependent decay (RIDD) and the ability to activate CD274 gene expression. Thus, the IRE1α/Xbp1 axis drives the polarization of macrophages in the tumor microenvironment initiating a complex immune dysregulation leading to failure of local immune surveillance.

Highlights

  • Myeloid cells in the tumor microenvironment (TME) are of central relevance to understand the dynamics of tumor progression [1]

  • Previous in vitro studies indicated that bone marrow–derived dendritic cell (BMDC) and bone marrow–derived macrophage (BMDM) respond to a cell-nonautonomous unfolded protein response (UPR) developing a complex phenotype characterized by a UPR activation and a mixed proinflammatory/immune suppressive (IIS) phenotype [26,28]

  • We implanted B16.F10 murine melanoma cells into C57BL/6 mice that carry the Xbp1-Venus fusion transgene under the control of the cytomegalovirus (CMV)-β actin promoter, known as the endoplasmic reticulum (ER) stress-activated indicator (ERAI) [41], which reports inositolrequiring enzyme 1 (IRE1α)-mediated X-box binding protein 1 (XBP1) splicing through the expression of the fluorescent Venus protein

Read more

Summary

Introduction

Myeloid cells in the tumor microenvironment (TME) are of central relevance to understand the dynamics of tumor progression [1]. Bone marrow–derived macrophages (BMDMs) and bone marrow–derived dendritic cells (BMDCs) cultured in conditioned media (CM) of ER stressed cancer cells develop a de novo UPR and acquire a mixed IIS phenotype [26,28] characterized by the transcriptional up-regulation of the tumorigenic proinflammatory cytokines IL-6, tumor necrosis factor α (TNFα), and IL-23 [32,33,34], and contextually of the immune-suppressive enzyme Arginase (Arg1) [35] Under these conditions, cross-priming of naïve CD8+ T cells by BMDC is greatly compromised [28]. We discovered that IRE1α signaling regulates programmed death ligand 1 (PD-L1) expression in murine and in tumor-infiltrating macrophages in humans

Results
Discussion
Materials and methods
Full Text
Published version (Free)

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