scholarly journals IRE1α regulates macrophage polarization, PD-L1 expression and tumor survival

2020 ◽  
Author(s):  
Alyssa Batista ◽  
Jeffrey J. Rodvold ◽  
Su Xian ◽  
Stephen Searles ◽  
Alyssa Lew ◽  
...  

ABSTRACTIn 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 upregulation of IL-6, IL-23, Arginase1, as well as surface expression of CD86 and PD-L1. Macrophages in which the IRE1α/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. 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.

Cancers ◽  
2020 ◽  
Vol 12 (5) ◽  
pp. 1288 ◽  
Author(s):  
Charlotte Dahlem ◽  
Wei Xiong Siow ◽  
Maria Lopatniuk ◽  
William K. F. Tse ◽  
Sonja M. Kessler ◽  
...  

Natural products represent powerful tools searching for novel anticancer drugs. Thioholgamide A (thioA) is a ribosomally synthesized and post-translationally modified peptide, which has been identified as a product of Streptomyces sp. MUSC 136T. In this study, we provide a comprehensive biological profile of thioA, elucidating its effects on different hallmarks of cancer in tumor cells as well as in macrophages as crucial players of the tumor microenvironment. In 2D and 3D in vitro cell culture models thioA showed potent anti-proliferative activities in cancer cells at nanomolar concentrations. Anti-proliferative actions were confirmed in vivo in zebrafish embryos. Cytotoxicity was only induced at several-fold higher concentrations, as assessed by live-cell microscopy and biochemical analyses. ThioA exhibited a potent modulation of cell metabolism by inhibiting oxidative phosphorylation, as determined in a live-cell metabolic assay platform. The metabolic modulation caused a repolarization of in vitro differentiated and polarized tumor-promoting human monocyte-derived macrophages: ThioA-treated macrophages showed an altered morphology and a modulated expression of genes and surface markers. Taken together, the metabolic regulator thioA revealed low activities in non-tumorigenic cells and an interesting anti-cancer profile by orchestrating different hallmarks of cancer, both in tumor cells as well as in macrophages as part of the tumor microenvironment.


Author(s):  
Chao Li

Endoplasmic reticulum (ER) stress triggers a series of signaling and transcriptional events termed the unfolded protein response (UPR). Severe ER stress is associated with the development of fibrosis in different organs including lung, liver, kidney, heart, and intestine. ER stress is an essential response of epithelial and immune cells in the pathogenesis of inflammatory bowel disease (IBD) including Crohn’s disease. Intestinal epithelial cells are susceptible to ER stress-mediated damage due to secretion of a large amount of proteins that are involved in mucosal defense. In other cells, ER stress is linked to myofibroblast activation, extracellular matrix production, macrophage polarization, and immune cell differentiation. This review focuses on the role of UPR in the pathogenesis in IBD from an immunologic perspective. The roles of macrophage and mesenchymal cells in the UPR from in vitro and in vivo animal models are discussed. The links between ER stress and other signaling pathways such as senescence and autophagy are introduced. Recent advances in the understanding of the epigenetic regulation of UPR signaling are also updated here. The future directions of development of the UPR research and therapeutic strategies to manipulate ER stress levels are also reviewed.


2019 ◽  
Vol 39 (10) ◽  
pp. 1967-1985 ◽  
Author(s):  
Hye Rim Chang ◽  
Tatjana Josefs ◽  
Diego Scerbo ◽  
Namrata Gumaste ◽  
Yunying Hu ◽  
...  

Objective: Fatty acid uptake and oxidation characterize the metabolism of alternatively activated macrophage polarization in vitro, but the in vivo biology is less clear. We assessed the roles of LpL (lipoprotein lipase)-mediated lipid uptake in macrophage polarization in vitro and in several important tissues in vivo. Approach and Results: We created mice with both global and myeloid-cell specific LpL deficiency. LpL deficiency in the presence of VLDL (very low-density lipoproteins) altered gene expression of bone marrow–derived macrophages and led to reduced lipid uptake but an increase in some anti- and some proinflammatory markers. However, LpL deficiency did not alter lipid accumulation or gene expression in circulating monocytes nor did it change the ratio of Ly6C high /Ly6C low . In adipose tissue, less macrophage lipid accumulation was found with global but not myeloid-specific LpL deficiency. Neither deletion affected the expression of inflammatory genes. Global LpL deficiency also reduced the numbers of elicited peritoneal macrophages. Finally, we assessed gene expression in macrophages from atherosclerotic lesions during regression; LpL deficiency did not affect the polarity of plaque macrophages. Conclusions: The phenotypic changes observed in macrophages upon deletion of Lpl in vitro is not mimicked in tissue macrophages.


2017 ◽  
Vol 114 (27) ◽  
pp. 7077-7082 ◽  
Author(s):  
Francesco Casciello ◽  
Fares Al-Ejeh ◽  
Greg Kelly ◽  
Donal J. Brennan ◽  
Shin Foong Ngiow ◽  
...  

G9a is an epigenetic regulator that methylates H3K9, generally causing repression of gene expression, and participates in diverse cellular functions. G9a is genetically deregulated in a variety of tumor types and can silence tumor suppressor genes and, therefore, is important for carcinogenesis. Although hypoxia is recognized to be an adverse factor in tumor growth and metastasis, the role of G9a in regulating gene expression in hypoxia has not been described extensively. Here, we show that G9a protein stability is increased in hypoxia via reduced proline hydroxylation and, hence, inefficient degradation by the proteasome. This inefficiency leads to an increase in H3K9me2 at its target promoters. Blocking the methyltransferase activity of G9a inhibited cellular proliferation and migration in vitro and tumor growth in vivo. Furthermore, an increased level of G9a is a crucial factor in mediating the hypoxic response by down-regulating the expression of specific genes, includingARNTL,CEACAM7,GATA2,HHEX,KLRG1, andOGN. This down-regulation can be rescued by a small molecule inhibitor of G9a. Based on the hypothesis that the changes in gene expression would influence patient outcomes, we have developed a prognostic G9a-suppressed gene signature that can stratify breast cancer patients. Together, our findings provide an insight into the role G9a plays as an epigenetic mediator of hypoxic response, which can be used as a diagnostic marker, and proposes G9a as a therapeutic target for solid cancers.


2021 ◽  
Vol 3 (1) ◽  
pp. 31-43
Author(s):  
Chao Li

Endoplasmic reticulum (ER) stress triggers a series of signaling and transcriptional events termed the unfolded protein response (UPR). Severe ER stress is associated with the development of fibrosis in different organs, including lung, liver, kidney, heart, and intestine. ER stress is an essential response of epithelial and immune cells in the pathogenesis of Inflammatory Bowel Disease (IBD), including Crohn’s disease (CD). Intestinal epithelial cells are susceptible to ER stress-mediated damage due to secretion of a large amount of proteins that are involved in mucosal defense. In other cells, ER stress is linked to myofibroblast activation, extracellular matrix production, macrophage polarization, and immune cell differentiation. This review focuses on the role of the UPR in the pathogenesis in IBD from an immunologic perspective. The roles of macrophage and mesenchymal cells in the UPR from in vitro and in vivo animal models are discussed. The links between ER stress and other signaling pathways, such as senescence and autophagy, are introduced. Recent advances in the understanding of the epigenetic regulation of the UPR signaling are also updated here. The future directions of development of the UPR research and therapeutic strategies to manipulate ER stress levels are also reviewed.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A745-A745
Author(s):  
Christie Mortales ◽  
Benjamin Dutzar ◽  
Jerry Chen ◽  
Alex Chen ◽  
Justin Huard ◽  
...  

BackgroundNL-201 is a potent, selective, and long-acting computationally designed alpha-independent agonist of the IL-2 and IL-15 receptors that is being developed as an immunotherapy for cancer. Downregulation of MHC class I (MHC-I) expression by tumors is a well-known mechanism of immune escape, and IFNγ is known to upregulate MHC-I. Here, we investigated whether NL-201 monotherapy can convert a 'cold' tumor microenvironment (TME) to an immunologically 'hot' TME through IFNγ-mediated MHC-I expression. This effect could expand the TCR repertoire for increased antitumor response and improve anti-PD-1 combination therapy.MethodsFor in vitro assays, mouse splenocytes were cultured with Neo-2/15 to assess effector cell function, as well as co-cultured with B16F10 cells to assess IFNγ-induced MHC-I and PD-L1 expression. B16F10 tumors were established in C57BL/6 mice and dosed with NL-201, anti-PD-1, or both to assess in vivo efficacy. B16F10 tumors were excised and dissociated for phenotyping of tumor-infiltrating lymphocytes (TILs) using flow cytometry. For gene expression analysis, RNA and genomic DNA were extracted from tumors and submitted for NanoString Pancancer Immune Profiling and Adaptive ImmunoSEQ analysis, respectively.ResultsIn vitro, Neo-2/15 induced greater CD8+ T cell and NK cell proliferation, as well as granzyme B production and IFNγ-dependent MHC-I upregulation on B16F10 tumor cells, compared to IL-2 or IL-15. In 'cold' B16F10 syngeneic tumors, NL-201 monotherapy reduced tumor growth and induced MHC-I, IFNγ, and granzyme B upregulation. Gene expression analysis of NL-201–treated tumors demonstrated increased TCR repertoire diversity and inflammatory signature at the tumor. In addition, PD-L1 was significantly upregulated on B16F10 cells. While the B16F10 tumors exhibited resistance to anti-PD-1 monotherapy, combination treatment with NL-201 significantly improved anti-PD-1 activity. This may explain the potent anti-tumor activity of NL-201 with anti-PD-1 combination therapy.ConclusionsNL-201 induces potent inflammatory effects on effector cells and is able to turn 'cold' TMEs 'hot'. We demonstrate that NL-201 strongly upregulated MHC-I expression in vitro and in vivo via an IFNγ-dependent pathway. Increased antigen presentation drives TCR diversity while augmenting the inflammatory signature at the tumor. This adaptive response also upregulates PD-L1 expression and results in impressive antitumor activity when NL-201 and PD-1 inhibitors are co-administered. The demonstration that NL-201 can convert 'cold' tumors to immunologically 'hot' tumors may provide a novel therapeutic option for patients unresponsive to current standard of care checkpoint inhibitors. A Phase 1 study of NL-201 in patients with advanced solid tumors is currently underway (NCT04659629).Ethics ApprovalAll experiments were approved by the Institutional Animal Care and Use Committee of Bloodworks Northwest and performed under protocol 5360-03.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A627-A627
Author(s):  
Cleopatra Rutihinda ◽  
Leanié Moreau ◽  
Safia Chelighem ◽  
Ayman Oweida

BackgroundRadiotherapy (RT) is commonly used to treat solid tumors but its efficacy varies widely. In non-virally driven head and neck cancer (HNC), radioresistance is responsible for most cases of tumor progression and tumor recurrence. The combination of RT and immunotherapy (IT), in the form of anti-PD-1/PD-L1, improved survival for a minority of patients with tumors that have pre-existing immunity or which are susceptible to RT-induced tumor immunity. In contrast, tumors with an immunosuppressive microenvironment show resistance to RT and/or IT. Regulatory T cells (Tregs) have been shown to be prevalent in HNC tumors and can promote treatment resistance. We identified the chemokine, CCL20 as a factor that can promote infiltration of Tregs into HNC tumors. CCL20 is secreted by the cancer cell and binds to its sole receptor, CCR6, expressed on Tregs. We therefore hypothesized that radiation induces CCL20 secretion resulting in infiltration of Tregs and radioresistance. We further hypothesized that blocking CCL20 in HNCs where CCL20 is induced in response to RT can decrease tumor growth.MethodsHuman and murine HNC cell lines (SCC9, Cal27, MOC1, MOC2) were irradiated at doses of 2Gy or 10Gy. Conditioned media (CM), RNA and protein were obtained 24h and 72h after radiation. The concentration of CCL20 was analyzed using a chemokine array. Gene expression was determined using qPCR. For in vivo studies, the MOC2 cell line was used. Mice were inoculated in the buccal cavity. Neutralizing CCL20 antibody was administered alone and in combination with RT. Blood samples were collected before and after RT for analysis of serum levels of CCL20.ResultsGene expression analysis showed that Cal27 and MOC2 tumors had a gene signature associated with immune-suppression and Treg cell infiltration. In contrast, SCC9 and MOC1 tumors displayed a gene signature associated with an inflamed microenvironment. In vitro, radiation induced a significant increase in CCL20 gene expression and secreted CCL20 in Cal27 and MOC2 cells relative to control. In contrast, MOC1 and SCC9 did not show a significant increase in CCL20 after radiation. In vivo, inhibition of CCL20 decreased tumor growth compared to control in MOC2 tumors. The combination of RT and anti-CCL20 showed a significant decrease in tumor growth compared to RT alone.ConclusionsCollectively, our data suggest that radiation promotes the induction of CCL20 in tumors with immune-suppressive mechanisms and inhibition of CCL20 can enhance the response to RT.Ethics ApprovalAll animal studies were conducted in accordance with the animal protection and ethics committee of the faculty of medicine at Universite de Sherbrooke (Le Comité facultaire de protection des animaux). Protocol #: 2019–2333.


2021 ◽  
Vol 11 ◽  
Author(s):  
Xuejin Zhu ◽  
Yangjia Zhuo ◽  
Shulin Wu ◽  
Yanfei Chen ◽  
Jianheng Ye ◽  
...  

Transcription factor EB (TFEB), a member of the MiT family, is dysregulated in different cancers and exerts specific biological functions within the tumor microenvironment. Downregulation of TFEB induces macrophage polarization in the TME and promotes tumor progression. However, the biological role and clinical significance of TFEB in prostate cancer (PCa) remain unknown. This study aimed to identify the role of TFEB in PCa and its potential clinical value. We explored TFEB expression in PCa using public databases and verified its prognostic value using immunohistochemistry in PCa tissue samples. The results revealed that TFEB expression was up-regulated in PCa tissues and was associated with cancer metastasis. Next, overexpression of TFEB promoted PCa cell malignant behavior in in vivo and in vitro experiments. RNA-sequencing and bioinformatics analysis showed high expression of TFEB promoted lysosomal biogenesis and knockdown of TFEB expression decreased the number of lysosomes. Furthermore, the ATP-binding cassette transporter A2 (ABCA2) was identified as a target gene of TFEB, which was verified using the cleavage under targets and release using nuclease (CUT&RUN) assay and qRT-PCR. Silencing of ABCA2 reduced lysosomal biogenesis and decreased matrix metalloproteinases expression, which reduced PCa cell invasion and migration in the tumor microenvironment. Our study suggests that TFEB promotes PCa progression by regulating ABCA2 through lysosomal biogenesis and may serve as a prognostic factor or as a potential therapeutic target of PCa.


2016 ◽  
Vol 113 (48) ◽  
pp. E7759-E7768 ◽  
Author(s):  
Stefani Spranger ◽  
Jason J. Luke ◽  
Riyue Bao ◽  
Yuanyuan Zha ◽  
Kyle M. Hernandez ◽  
...  

Melanoma metastases can be categorized by gene expression for the presence of a T-cell–inflamed tumor microenvironment, which correlates with clinical efficacy of immunotherapies. T cells frequently recognize mutational antigens corresponding to nonsynonymous somatic mutations (NSSMs), and in some cases shared differentiation or cancer–testis antigens. Therapies are being pursued to trigger immune infiltration into non–T-cell–inflamed tumors in the hope of rendering them immunotherapy responsive. However, whether those tumors express antigens capable of T-cell recognition has not been explored. To address this question, 266 melanomas from The Cancer Genome Atlas (TCGA) were categorized by the presence or absence of a T-cell–inflamed gene signature. These two subsets were interrogated for cancer–testis, differentiation, and somatic mutational antigens. No statistically significant differences were observed, including density of NSSMs. Focusing on hypothetical HLA-A2+binding scores, 707 peptides were synthesized, corresponding to all identified candidate neoepitopes. No differences were observed in measured HLA-A2 binding between inflamed and noninflamed cohorts. Twenty peptides were randomly selected from each cohort to evaluate priming and recognition by human CD8+T cells in vitro with 25% of peptides confirmed to be immunogenic in both. A similar gene expression profile applied to all solid tumors of TCGA revealed no association between T-cell signature and NSSMs. Our results indicate that lack of spontaneous immune infiltration in solid tumors is unlikely due to lack of antigens. Strategies that improve T-cell infiltration into tumors may therefore be able to facilitate clinical response to immunotherapy once antigens become recognized.


Author(s):  
Chao Li

Endoplasmic reticulum (ER) stress triggers a series of signaling and transcriptional events termed the unfolded protein response (UPR). Severe ER stress is associated with the development of fibrosis in different organs including lung, liver, kidney, heart, and intestine. ER stress is an essential response of epithelial and immune cells in the pathogenesis of inflammatory bowel disease (IBD) including Crohn’s disease. Intestinal epithelial cells are susceptible to ER stress-mediated damage due to secretion of a large amount of proteins that are involved in mucosal defense. In other cells, ER stress is linked to myofibroblast activation, extracellular matrix production, macrophage polarization, and immune cell differentiation. This review focuses on the role of UPR in the pathogenesis in IBD from immunologic perspective. The roles of macrophage and mesenchymal cells in the UPR from in vitro and in vivo animal models are discussed. The links between ER stress and other signaling pathways such as senescence and autophagy are introduced. Recent advances in the understanding of the epigenetic regulation of UPR signaling are reported. The future directions of the development of the UPR research and therapeutic strategies to manipulate ER stress levels are also reviewed.


Sign in / Sign up

Export Citation Format

Share Document