Abstract PO021: Lung cancer cells and cancer-associated fibroblasts drive macrophage polarization in a co-culture model

Author(s):  
Josiah Flaming ◽  
Raghav Chandra ◽  
Luc Girard ◽  
Debolina Ganguly ◽  
Jason Toombs ◽  
...  
2017 ◽  
Vol 7 (1) ◽  
Author(s):  
Ryu Kanzaki ◽  
Hisamichi Naito ◽  
Kazuyoshi Kise ◽  
Kazuhiro Takara ◽  
Daisuke Eino ◽  
...  

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A793-A794
Author(s):  
Josiah Flaming ◽  
Raghav Chandra ◽  
Luc Girard ◽  
Debolina Ganguly ◽  
Jason Toombs ◽  
...  

BackgroundThe plasticity of macrophage phenotype within the tumor microenvironment (TME) correlates with prognosis in non-small cell lung cancer (NSCLC).1 M2-like macrophages promote immunosuppression and facilitate tumor progression, while M1-like macrophages may drive an inflammatory antitumor immune response.2 Through a novel co-culture model comprised of cancer cells, cancer-associated fibroblasts (CAFs), and macrophages, we investigated whether NSCLC oncogenotype impacts macrophage phenotype and postulated that the immunosuppressive activity of macrophages is mediated through tumor-secreted soluble molecules. If identified and inhibited, these may re-sensitize cancer cells to immune surveillance and enhance antitumor immunity.MethodsWe developed an in vitro co-culture system (patient-derived NSCLC cells, human CAFs, and mouse macrophages) to interrogate impact of NSCLC cells and CAFs on macrophage phenotype. Expression of salient macrophage genes (i.e. ARG1, NOS2, IL-1β, IL-6, CHIL-3, SOCS3) was investigated through species-specific qPCR. Whole-genome RNA sequencing (RNAseq) in select cases was conducted and cytokine arrays measuring expression of 40 inflammatory cytokines were performed. Positive controls included stimulation of macrophages with LPS and IL-4.ResultsMore than 70 NSCLC cell lines were characterized in the co-culture assay. Three highly reproducible clusters of macrophage phenotypes were identified: high Arginase (immunosuppressive), high IL-1β (inflammatory) and high SOCS3 (inflammatory, involved in JAK-STAT3 pathway) (figure 1).3 4 Major oncogenotypes (i.e. KRAS, TP53, STK11, EGFR, BRAF mutation) did not correlate with macrophage phenotype (figure 2). Analyses of differences between the 3 clusters is ongoing. 10 exemplar NSCLC lines representing each of these 3 clusters were selected for RNA sequencing (mouse genes) and cytokine array protein (human) profiling. Across all clusters, we found suppression of macrophage endocytosis pathways and activation of scavenger receptor A (SRA) signaling, reflecting an M2-like phenotype.5 We also observed increased expression of human IL-6, IL-8, and MCP1, which are implicated in suppression of innate immune sensing of tumor cells (figure 3). RNAseq of CAF lines demonstrated mixed inflammatory and myofibroblastic phenotypes (figure 4), with increased expression of genes associated with macrophage recruitment and activation including: IL-6, CSF-1, CXCL6, CCL2, and CCL7.6Abstract 746 Figure 1Three macrophage phenotypes induced in co-cultureHeatmap of mRNA expression from mouse macrophages co-cultured with human NSCLC cells and CAFs. mRNA expression of salient mouse macrophage genes depicted (x-axis) for each NSCLC cell line co-culture (y-axis).Abstract 746 Figure 2Macrophage phenotype independent of oncogenotypePercentage of mutations of known human NSCLC oncogenes per mouse macrophage phenotype cluster.Abstract 746 Figure 3Upregulation of macrophage-related cytokinesCytokine array assays demonstrating relative expression of cytokines and chemokines from individual cell types or multicellular co-cultures associated with macrophage recruitment and polarizationAbstract 746 Figure 4Mixed expression of iCAF and myCAF genes on RNAseqHeatmap of RNAseq transcriptome of human CAFs from co-culture model reflecting relative expression of known genes associated inflammatory (iCAF, top) and myofibroblastic (myCAF, bottom) phenotypes.Abstract 746 Figure 5Novel co-culture model of NSCLC TMEDepiction of novel co-culture model with mouse bone-marrow derived macrophages, human NSCLC cells, and human CAFs with a representative immunohistochemical fluorescence image in vitroConclusionsThrough this novel co-culture model (figure 5), we demonstrate that patient-derived NSCLC cells reproducibly induce three major macrophage phenotypes in an oncotype-independent manner. Furthermore, cytokine release from NSCLC cells and CAFs is implicated in this process. This co-culture model provides a physiologically consistent experimental platform to identify tumor cell and CAF features that drive macrophage phenotype which may be suitable for targeted therapy.AcknowledgementsWe thank the McDermott Center Next-Generation Sequencing Core at UT Southwestern. Figure 5 was created with Biorender.comReferencesSumitomo R, Hirai T, Fujita M, et al. M2 tumor associated macrophages promote tumor progression in non small cell lung cancer. Exp Ther Med 2019 Dec 1;18(6):4490–8.Chen Y, Song Y, Du W, et al. Tumor-associated macrophages: an accomplice in solid tumor progression. J. Biomed. Sci 2019 Dec;26(1):1–3.Orecchioni M, Ghosheh Y, Pramod A, et al. Macrophage polarization: different gene signatures in M1 (LPS+) vs. classically and M2 (LPS–) vs. alternatively activated macrophages. Front. Immunol 2019 May 24;10:1084.Wilson HM. SOCS proteins in macrophage polarization and function. Front. Immunol 2014 Jul 28;5:357.Sun Y, Xu S. Tumor-associated CD204-positive macrophage is a prognostic marker in clinical stage I lung adenocarcinoma. Biomed Res. Int 2018 Jan 1;2018.O’Hayre M, Salanga C, Handel T, et al. Chemokines and cancer: migration, intracellular signalling and intercellular communication in the microenvironment. Biochem. J 2008 Feb 1;409(3):635–49


RSC Advances ◽  
2016 ◽  
Vol 6 (29) ◽  
pp. 24083-24090 ◽  
Author(s):  
Dan-Dan Wang ◽  
Wei Liu ◽  
Jing-Jie Chang ◽  
Xu Cheng ◽  
Xiu-Zhen Zhang ◽  
...  

Bioengineering a three-dimensional culture model of human lung cancer cells for screening EGFR targeted inhibitors.


QJM ◽  
2019 ◽  
Vol 112 (8) ◽  
pp. 581-590 ◽  
Author(s):  
J You ◽  
M Li ◽  
L M Cao ◽  
Q H Gu ◽  
P B Deng ◽  
...  

Abstract Background Epithelial-mesenchymal transition (EMT) is an essential component of metastasis. Our previous study demonstrated that cancer-associated fibroblasts (CAFs) induce EMT in lung cancer cells. In recent years, many studies have demonstrated that CAFs induce metastasis and drug resistance in cancer cells via exosomes. Aim We sought to discover the mechanism underlying how CAFs induce EMT in lung cancer cells, unveiling the role of exosomes in lung cancer progression. Design We cultured lung cancer cell (i) with control medium, normal fibroblasts (NFs) or CAFs; (ii) with SNAI1-transfected or NC (negative control)-transfected CAFs; (iii) with exosomes extracted from NF- or CAF-conditioned medium; (iv) with exosomes released by SNAI1 or NC-transfected CAFs; (v) with CAF-conditioned medium or exosome-depleted CAF-conditioned medium. Methods qRT-PCR was conducted to examine the expression of CDH1 (gene of E-cadherin) and VIM (gene of Vimentin), western blotting was conducted to examine E-cadherin and vimentin levels in lung cancer cells. Results Exosomes released by CAFs-promoted EMT in lung cancer cells. Interestingly, SNAI1 levels in exosomes secreted from CAFs were correlated with SNAI1 expression in CAFs. Furthermore, the level of SNAI1 in exosomes was crucial for inducing EMT in lung cancer cells. Finally, treatment of CAFs with GW4869, an inhibitor of exosome release, noticeably inhibited their EMT-inducing effect on recipient epithelial cells. Conclusions The molecular mechanism underlying how CAFs induce EMT in cancer cells may be that CAFs deliver SNAI1 to recipient cancer cells via exosomes.


2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Meijuan Chen ◽  
Cheng Hu ◽  
Qian Gao ◽  
Liqiu Li ◽  
Ziyu Cheng ◽  
...  

Abstract Background Kejinyan decoction, as an experienced formula of Zhou Zhongying (the Master of Traditional Chinese Medicine) has been widely used in clinic for lung cancer treatment in China, while the anti-lung cancer mechanism of it is still remained to be elucidated. Herein, our basic study found that the survival of lung cancer xenograft mice was significantly prolonged after intragastrically administered high dose of Kejinyan decoction (3.8 g per kg BW) for 15 days. More importantly, we found that Kejinyan decoction inhibited the metastasis of lung cancer cells in vivo. Thus in this study, we aim to elucidate the anti-metastasis effects of Kejinyan decoction. Methods RNA-Seq was used to find out the gene regulation of Kejinyan decoction on the mice, flow cytometry assay was used to detect the immunocytes in the spleen, ELISA assay was used to detect the inflammatory factors in the serum and spleen, and immunofluorescence assay was used to detect the level of immune cells and the expression of glycol-metabolism related enzymes in situ. Also, we established a lung cancer orthotopic xenograft tumor model to assess the influence of Kejinyan decoction on the metastatic ability of lung cancer cells in vivo. Results GO analysis of gene sequencing of tumor tissue samples showed that Kejinyan decoction regulated immune response. Further flow cytometry analysis of splenic lymphocyte showed that Kejinyan decoction upregulated M1 macrophages and downregulated M2 macrophages, while the total level of macrophages changed little, which was verified by detection of CD68, F4/80, CD206, and CD86 in tumor tissue section. Moreover, detection of inflammatory cytokines showed that Kejinyan decoction downregulated TNF-α, IFN-γ, IL-6, as well as IL-4, IL-13 in tumor microenvironment. Further studies also showed that Kejinyan decoction had little effect on tumor hypoxia, but downregulated glycolysis in tumor tissues. More importantly, we found that Kejinyan decoction inhibited the metastasis of lung cancer cells in vivo. Conclusion Our findings conclude that Kejinyan decoction inhibited lung cancer cell metastasis through affecting macrophage polarization and energy reprogramming.


2021 ◽  
Author(s):  
Sieun Lee ◽  
Ji Hyung Hong ◽  
Jeong Seon Kim ◽  
Jung Sook Yoon ◽  
Sang Hoon Chun ◽  
...  

Author(s):  
Xueru Guo ◽  
Mengmeng Chen ◽  
Limin Cao ◽  
Yiming Hu ◽  
Xueqin Li ◽  
...  

Cancer-associated fibroblasts (CAFs) are major component of tumor microenvironment (TME), which plays crucial roles in tumor growth, invasion and metastasis; however, the underling mechanism is not fully elucidated. Despite many studies are focused on the tumor promoting effect of CAFs-derived cytokines, the upstream regulators of cytokine release in CAFs is largely unknown. Here we found that miR-101-3p was downregulated in primary lung cancer-associated CAFs compared to normal fibroblasts (NFs). Ectopic overexpression of miR-101-3p suppressed CAFs activation, and abrogated the promoting effect of CAFs on migration and invasion of non-small cell lung cancer cells (NSCLC), through attenuating CAFs’ effect on epithelial mesenchymal transition (EMT) process, metastasis-related genes (MMP9, TWIST1) and AKT/endothelial nitric oxide synthase (eNOS) signaling pathway. Further study indicated that vascular endothelial growth factor A (VEGFA) was a novel target of miR-101-3p, and CAFs-derived VEGFA mediated the effect of miR-101-3p on migration and invasion of lung cancer cells, demonstrated by using recombinant VEGFA and VEGFA neutralizing antibody. Interestingly, the analysis of the Cancer Genome Atlas (TCGA) database revealed that lung cancer tissues expressed lower level of miR-101-3p than non-cancerous tissues, and low/medium-expression of miR-101-3p was associated with poor overall survival (OS) rate. Moreover, the mouse xenograft experiment also showed that CAFs accelerated tumor growth whereas miR-101-3p diminished CAFs’ effect. These findings revealed a novel mechanism that CAFs facilitated lung cancer metastasis potential via miR-101-3p/VEGFA/AKT signaling pathway, suggesting miR-101-3p as a potential candidate for metastasis therapy.


Sign in / Sign up

Export Citation Format

Share Document