Exosomal circPVT1 derived from lung cancer promotes the progression of lung cancer by targeting miR-124-3p/EZH2 axis and regulating macrophage polarization

Cell Cycle ◽  
2022 ◽  
pp. 1-17
Author(s):  
Ying Liu ◽  
Lei Li ◽  
Xiang Song
2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Theodora Katopodi ◽  
Savvas Petanidis ◽  
Kalliopi Domvri ◽  
Paul Zarogoulidis ◽  
Doxakis Anestakis ◽  
...  

AbstractIntratumoral heterogeneity in lung cancer is essential for evasion of immune surveillance by tumor cells and establishment of immunosuppression. Gathering data reveal that circular RNAs (circRNAs), play a role in the pathogenesis and progression of lung cancer. Particularly Kras-driven circRNA signaling triggers infiltration of myeloid-associated tumor macrophages in lung tumor microenvironment thus establishing immune deregulation, and immunosuppression but the exact pathogenic mechanism is still unknown. In this study, we investigate the role of oncogenic Kras signaling in circRNA-related immunosuppression and its involvement in tumoral chemoresistance. The expression pattern of circRNAs HIPK3 and PTK2 was determined using quantitative polymerase chain reaction (qPCR) in lung cancer patient samples and cell lines. Apoptosis was analyzed by Annexin V/PI staining and FACS detection. M2 macrophage polarization and MDSC subset analysis (Gr1−/CD11b−, Gr1−/CD11b+) were determined by flow cytometry. Tumor growth and metastatic potential were determined in vivo in C57BL/6 mice. Findings reveal intra-epithelial CD163+/CD206+ M2 macrophages to drive Kras immunosuppressive chemoresistance through myeloid differentiation. In particular, monocytic MDSC subsets Gr1−/CD11b−, Gr1−/CD11b+ triggered an M2-dependent immune response, creating an immunosuppressive tumor-promoting network via circHIPK3/PTK2 enrichment. Specifically, upregulation of exosomal cicHIPK3/PTK2 expression prompted Kras-driven intratumoral heterogeneity and guided lymph node metastasis in C57BL/6 mice. Consequent co-inhibition of circPTK2/M2 macrophage signaling suppressed lung tumor growth along with metastatic potential and prolonged survival in vivo. Taken together, these results demonstrate the key role of myeloid-associated macrophages in sustaining lung immunosuppressive neoplasia through circRNA regulation and represent a potential therapeutic target for clinical intervention in metastatic lung cancer.


2021 ◽  
Vol 22 (2) ◽  
pp. 746
Author(s):  
Radu Pirlog ◽  
Andrei Cismaru ◽  
Andreea Nutu ◽  
Ioana Berindan-Neagoe

Lung cancer is currently the first cause of cancer-related death. The major lung cancer subtype is non-small cell lung cancers (NSCLC), which accounts for approximatively 85% of cases. The major carcinogenic associated with lung cancer is tobacco smoke, which produces long-lasting and progressive damage to the respiratory tract. The progressive and diffuse alterations that occur in the respiratory tract of patients with cancer and premalignant lesions have been described as field cancerization. At the level of tumor cells, adjacent tumor microenvironment (TME) and cancerized field are taking place dynamic interactions through direct cell-to-cell communication or through extracellular vesicles. These molecular messages exchanged between tumor and nontumor cells are represented by proteins, noncoding RNAs (ncRNAs) and microRNAs (miRNAs). In this paper, we analyze the miRNA roles in the macrophage polarization at the level of TME and cancerized field in NSCLC. Identifying molecular players that can influence the phenotypic states at the level of malignant cells, tumor microenvironment and cancerized field can provide us new insights into tumor regulatory mechanisms that can be further modulated to restore the immunogenic capacity of the TME. This approach could revert alterations in the cancerized field and could enhance currently available therapy approaches.


2018 ◽  
Vol 47 (6) ◽  
pp. 2534-2543 ◽  
Author(s):  
Changjun He ◽  
Kaibin Zhu ◽  
Xue Bai ◽  
Yingbin Li ◽  
Dawei Sun ◽  
...  

Background/Aims: Assistance with tumor-associated vascularization is needed for the growth and invasion of non-small cell lung cancer (NSCLC). Recently, it was shown that placental growth factor (PLGF) expressed by NSCLC cells had a critical role in promoting the metastasis of NSCLC cells. However, the underlying molecular mechanisms remain elusive. Methods: Here, we first established a NSCLC model in mice that allows us not only to isolate tumor cells from non-tumor cells in the tumor, but also to trace tumor cells in living animals. Levels of PLGF, its unique receptor Flt-1, as well as transforming growth factor β1 (TGFβ1) was examined in tumor cells and tumor-associated macrophages (TAM) by RT-qPCR. A transwell well co-culture system and HUVEC assay were applied to study the crosstalk between NSCLC cells and TAM. Results: NSCLC cells produced and secreted PLGF to signal to tumor-associated macrophages (TAM) through surface expression of Flt-1 on macrophages. In a transwell co-culture system, PLGF secreted by NSCLC cells triggered macrophage polarization to a TAM subtype that promote growth of NSCLC cells. Moreover, polarized TAM seemed to secrete TGFβ1 to enhance the growth of endothelial cells in a HUVEC assay. Conclusion: The cross-talk between TAM and NSCLC cells via PLGF/Flt-1 and TGFβ receptor signaling may promote the growth and vascularization of NSCLC.


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


Author(s):  
Iwona Osinska ◽  
Dominika Wolosz ◽  
Magorzata Proboszcz ◽  
Dariusz Dziedzic ◽  
Magorzata Polubiec-Kownacka ◽  
...  

2021 ◽  
Vol 11 ◽  
Author(s):  
Kayla F. Goliwas ◽  
Hannah M. Ashraf ◽  
Anthony M. Wood ◽  
Yong Wang ◽  
Kenneth P. Hough ◽  
...  

Tumor-stromal interactions within the tumor microenvironment (TME) influence lung cancer progression and response to therapeutic interventions, yet traditional in vitro studies fail to replicate the complexity of these interactions. Herein, we developed three-dimensional (3D) lung tumor models that mimic the human TME and demonstrate tumor-stromal crosstalk mediated by extracellular vesicles (EVs). EVs released by tumor cells, independent of p53 status, and fibroblasts within the TME mediate immunomodulatory effects; specifically, monocyte/macrophage polarization to a tumor-promoting M2 phenotype within this 3D-TME. Additionally, immune checkpoint inhibition in a 3D model that included T cells showed an inhibition of tumor growth and reduced hypoxia within the TME. Thus, perfused 3D tumor models incorporating diverse cell types provide novel insights into EV-mediated tumor-immune interactions and immune-modulation for existing and emerging cancer therapies.


Sign in / Sign up

Export Citation Format

Share Document