Glioma exosomes mediate the expansion and function of myeloid-derived suppressor cells through microRNA-29a/Hbp1 and microRNA-92a/Prkar1a pathways
Xiaofan Guo
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
First author's name: Xiaofan Guo and Wei Qiu.Search for more papers by this authorWei Qiu
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
First author's name: Xiaofan Guo and Wei Qiu.Search for more papers by this authorJian Wang
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
Department of Biomedicine, University of Bergen, Bergen, Norway
Search for more papers by this authorQinglin Liu
Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorMingyu Qian
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorShaobo Wang
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorZongpu Zhang
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorXiao Gao
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorZihang Chen
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorQindong Guo
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorJianye Xu
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorCorresponding Author
Hao Xue
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
Correspondence to: Gang Li, Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Tel.: 086-0531-82166615, Fax: 086-0531-82166615, E-mail: [email protected]; or Hao Xue, Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, E-mail: [email protected]Search for more papers by this authorCorresponding Author
Gang Li
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
Correspondence to: Gang Li, Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Tel.: 086-0531-82166615, Fax: 086-0531-82166615, E-mail: [email protected]; or Hao Xue, Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, E-mail: [email protected]Search for more papers by this authorXiaofan Guo
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
First author's name: Xiaofan Guo and Wei Qiu.Search for more papers by this authorWei Qiu
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
First author's name: Xiaofan Guo and Wei Qiu.Search for more papers by this authorJian Wang
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
Department of Biomedicine, University of Bergen, Bergen, Norway
Search for more papers by this authorQinglin Liu
Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorMingyu Qian
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorShaobo Wang
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorZongpu Zhang
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorXiao Gao
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorZihang Chen
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorQindong Guo
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorJianye Xu
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Search for more papers by this authorCorresponding Author
Hao Xue
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
Correspondence to: Gang Li, Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Tel.: 086-0531-82166615, Fax: 086-0531-82166615, E-mail: [email protected]; or Hao Xue, Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, E-mail: [email protected]Search for more papers by this authorCorresponding Author
Gang Li
Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, People's Republic of China
Shandong Provincial Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, People's Republic of China
Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
Correspondence to: Gang Li, Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Tel.: 086-0531-82166615, Fax: 086-0531-82166615, E-mail: [email protected]; or Hao Xue, Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, E-mail: [email protected]Search for more papers by this authorAbstract
Myeloid-derived suppressor cells (MDSCs) play a pivotal role in mediating the formation of an immunosuppressive environment and assisting tumors in evading the host immune response. However, the mechanism through which tumors manipulate the differentiation and function of MDSCs remains unclear. Here, we report that hypoxia-induced glioma cells can stimulate the differentiation of functional MDSCs by transferring exosomal miR-29a and miR-92a to MDSCs. Our results showed that glioma-derived exosomes (GEXs) can enhance the differentiation of functional MDSCs both in vitro and in vivo, and hypoxia-induced GEXs (H-GEXs) demonstrated a stronger MDSCs induction ability than did normoxia-induced GEXs (N-GEXs). A subsequent miRNA sequencing analysis of N-GEXs and H-GEXs revealed that hypoxia-induced exosomal miR-29a and miR-92a expression induced the propagation of MDSCs. miR-29a and miR-92a activated the proliferation and function of MDSCs by targeting high-mobility group box transcription factor 1 (Hbp1) and protein kinase cAMP-dependent type I regulatory subunit alpha (Prkar1a), respectively. Altogether, the results of our study provide new insights into the role of glioma exosomal miRNAs in mediating the formation of immunosuppressive microenvironments in tumors and elucidate the underlying exosomal miR-29a/miR-92a-based regulatory mechanism responsible for the modulation of functional MDSC induction.
Abstract
What's new?
Myeloid derived suppressor cells (MDSC) suppress the immune system, reducing the effectiveness of immunotherapy in various cancers. These authors investigated how hypoxia promotes the immunosuppressive activity of MDSCs. Glioma cells, they found, use exosomes to transport miRNAs to MDSC progenitor cells, stimulating their differentiation. Hypoxia-induced exosomes exerted a more powerful effect on MDSC proliferation than normoxia-induced exosomes. The miRNAs induced MDSCs by targeting two genes, Hbp1 and Prkar1a. miRNA silencing of Hbp1 induced the cell cycle progression of MDSCs. This is the first study to demonstrate a mechanism by which glioma cells stimulate MDSC expansion.
Supporting Information
Filename | Description |
---|---|
ijc32052-sup-0001-appendixS1.docWord document, 35 KB | Appendix S1: Supplemental Methods. |
ijc32052-sup-0002-FigureS1.tifTIFF image, 5.2 MB |
Supplementary Figure S1 Exosomes secreted by glioma cells are internalized by MDSCs A. Electron microscopy images of exosomes secreted by different glioma cell lines stimulated with normoxia or hypoxia (N-GEXs or H-GEXs). Scale bar: 100 nm. B. Particle diameter and concentration analysis of GEXs measured by qNano. C. Western blotting analysis of GEXs and cells for the negative exosome marker Calnexin and the positive exosome markers TSG101 and CD9. D. Confocal microscopy images of MDSCs incubated with PKH67 (green)-labeled GEXs derived from the P3 and GL261 glioma cell lines for 0, 6, 24 and 48 h. Phalloidin-TRITC (red) and DAPI (blue) were used to stain MDSCs. Scale bar: 5 μm. E. Bioluminescence imaging confirmed glioma formation, and representative confocal microscopy images validated the internalization of GEXs by MDSCs in vivo at 2 or 4 weeks after the implantation of recombinase-expressing GL261 cells. MDSCs in non-injected mice or mice implanted with normal GL261 cells were used as controls. Scale bar: 100 μm. The percentage of positive cells (ratio of the number of cells expressing eGFP to the total number of cells observed) was calculated. NS, not significant. F. The bone marrow cells were treated with PKH67 labeled H-GEXs derived from GL261 and prepared for flow cytometry 7 days later. The bone marrow cells that have taken the PKH67 labeled exosomes were shown (F). The percentage of MDSCs in bone marrow cells have taken PKH67 labeled exosomes (P2) or not (P1) were shown (G). The percentage of MDSCs in all bone marrow cells was also shown (P1 and P2). *P < 0.05, **P < 0.01, ***P < 0.005, NS, not significant. (n = 3; mean ± s.e.m) |
ijc32052-sup-0003-FigureS2.tifTIFF image, 869 KB |
Supplementary Figure S2 miRNA expression in GEXs A. The supernatants of normoxic GL261/G422 cells transfected with control, miR-29a-overexpressing or miR-92a-overexpressing lentiviruses and hypoxic GL261/G422 cells transfected with control, miR-29a-knockout or miR-92a-knockout lentiviruses were collected to isolate exosomes (N-GEXs, N + 29 GEXs, N + 92 GEXs, H-GEXs, H-29KO GEXs, and H-92KO GEXs). Supernatants of hypoxic GL261/G422 cells transfected miR-29a and miR-92a knockout lentiviruses together were also collected to isolate exosomes (H-29-92 double KO GEXs). The miR-29a and miR-92a miRNA expression in these GEXs was detected by qRT-PCR. B. Normal or Cre recombinase-expressing GL261 cells were cultured for 48 h, and the culture medium was used to isolate exosomes. The Cre mRNA expression in these GEXs was detected by qRT-PCR. *P < 0.05, **P < 0.01, ***P < 0.005, NS, not significant. (n = 3; mean ± s.e.m) |
ijc32052-sup-0004-FigureS3.tifTIFF image, 496.9 KB |
Supplementary Figure S3 The expression of miR-29a and miR-92a in splenic MDSCs The expression of miR-29a and miR-92a in splenic MDSCs isolated from glioma bearing mouse (the mouse was intracranially injected with GL261 glioma cells one months ago) and control mouse (the mouse was intracranially injected with PBS one months ago) was detected by qRT-PCR. *P < 0.05, **P < 0.01, ***P < 0.005, NS, not significant. (n = 3; mean ± s.e.m) |
ijc32052-sup-0005-FigureS4.tifTIFF image, 639.6 KB |
Supplementary Figure S4 Optiprep density gradient. Hypoxia induced exosomes derived from U87 cells were isolated using an iodixanol density gradient centrifugation. Fractions were subjected to qPCR analysis with primers specific to miR-29a and miR-92a as well as to western blot analysis with the exosome marker protein CD9. |
ijc32052-sup-0006-FigureS5.tifTIFF image, 1.7 MB |
Supplementary Figure S5 siRNA, mimic, antagomirs and PCR primer sequences A. Sequences of Hbp1, Prkar1a and control siRNAs. B. Sequences of miR-29a-3p mimics, miR-92a-3p mimics, miR-29a-3p antagomir, miR-92a-3p antagomir and control. C. Sequences of PCR primers. |
ijc32052-sup-0007-FigureS6.tifTIFF image, 3.9 MB | Supplementary Figure S6 The uncropped scans of western blots from Figure 6 |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
References
- 1Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009; 9: 162–74.
- 2McNeill KA. Epidemiology of brain tumors. Neurol Clin 2016; 34: 981–98.
- 3Rodrigues JC, Gonzalez GC, Zhang L, et al. Normal human monocytes exposed to glioma cells acquire myeloid-derived suppressor cell-like properties. Neuro Oncol 2010; 12: 351–65.
- 4van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. LID - 10.1038/nrm.2017.125 [doi].
- 5Nawaz M, Fatima F. Extracellular vesicles, tunneling nanotubes, and cellular interplay: synergies and missing links. Front Mol Biosci 2017; 4: 50.
- 6Mateescu B, Kowal EJ, van Balkom BW, et al. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA - an ISEV position paper. J Extracell Vesicles 2017; 6:1286095.
- 7Nawaz M. Extracellular vesicle-mediated transport of non-coding RNAs between stem cells and cancer cells: implications in tumor progression and therapeutic resistance. Stem Cell Invest 2017; 4: 83.
- 8Xiang X, Poliakov A, Liu C, et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer 2009; 124: 2621–33.
- 9Fatima F, Nawaz M. Vesiculated long non-coding RNAs: offshore packages deciphering trans-regulation between cells, cancer progression and resistance to therapies. Noncoding RNA 2017; 3: 10.
- 10Valadi H, Ekstrom K, Bossios A, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007; 9: 654–9.
- 11Li L, Zhang J, Diao W, et al. MicroRNA-155 and MicroRNA-21 promote the expansion of functional myeloid-derived suppressor cells. J Immunol 2014; 192: 1034–43.
- 12Tadokoro H, Umezu T, Ohyashiki K, et al. Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. J Biol Chem 2013; 288: 34343–51.
- 13Rong Y, Durden DL, Van Meir EG, et al. 'Pseudopalisading' necrosis in glioblastoma: a familiar morphologic feature that links vascular pathology, hypoxia, and angiogenesis. J Neuropathol Exp Neurol 2006; 65: 529–39.
- 14Zomer A, Steenbeek SC, Maynard C, et al. Studying extracellular vesicle transfer by a Cre-loxP method. Nat Protoc 2016; 11: 87–101.
- 15Ridder K, Sevko A, Heide J, et al. Extracellular vesicle-mediated transfer of functional RNA in the tumor microenvironment. Oncoimmunology 2015; 4: e1008371.
- 16Muzumdar MD, Tasic B, Miyamichi K, et al. A global double-fluorescent Cre reporter mouse. Genesis 2007; 45: 593–605.
- 17Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol 2011; 32: 19–25.
- 18Berasi SP, Xiu M, Yee AS, et al. HBP1 repression of the p47phox gene: cell cycle regulation via the NADPH oxidase. Mol Cell Biol 2004; 24: 3011–24.
- 19Xiu M, Kim J, Sampson E, et al. The transcriptional repressor HBP1 is a target of the p38 mitogen-activated protein kinase pathway in cell cycle regulation. Mol Cell Biol 2003; 23: 8890–901.
- 20Sampson EM, Haque ZK, Ku MC, et al. Negative regulation of the Wnt-beta-catenin pathway by the transcriptional repressor HBP1. EMBO J 2001; 20: 4500–11.
- 21Bott A, Erdem N, Lerrer S, et al. miRNA-1246 induces pro-inflammatory responses in mesenchymal stem/stromal cells by regulating PKA and PP2A. Oncotarget 2017;8:43897–914.
- 22Yang Y, Pan X, Lei W, et al. Regulation of transforming growth factor-beta 1-induced apoptosis and epithelial-to-mesenchymal transition by protein kinase a and signal transducers and activators of transcription 3. Cancer Res 2006; 66: 8617–24.
- 23Noman MZ, Hasmim M, Messai Y, et al. Hypoxia: a key player in antitumor immune response. A review in the theme: cellular responses to hypoxia. Am J Physiol Cell Physiol 2015; 309: C569–79.
- 24Noman MZ, Janji B, Hu S, et al. Tumor-promoting effects of myeloid-derived suppressor cells are potentiated by hypoxia-induced expression of miR-210. Cancer Res 2015; 75: 3771–87.
- 25Kumar V, Patel S, Tcyganov E, et al. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol 2016;37:208–20.
- 26Whiteside TL. The effect of tumor-derived exosomes on immune regulation and cancer immunotherapy. Future Oncol 2017; 13: 2583–92.
- 27Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol 2014; 14: 195–208.
- 28Kunigelis KE, Graner MW. The Dichotomy of Tumor Exosomes (TEX) in cancer immunity: Is It All in the ConTEXt? Vaccines (Basel) 2015; 3(4): 1019–51.
- 29Liu Y, Gu Y, Cao X. The exosomes in tumor immunity. Oncoimmunology 2015; 4(9): e1027472.
- 30Dai S, Zhou X, Wang B, et al. Enhanced induction of dendritic cell maturation and HLA-A*0201-restricted CEA-specific CD8(+) CTL response by exosomes derived from IL-18 gene-modified CEA-positive tumor cells. J Mol Med (Berl) 2006; 84: 1067–76.
- 31Bollaert E, Johanns M, Herinckx G, et al. HBP1 phosphorylation by AKT regulates its transcriptional activity and glioblastoma cell proliferation. Cell Signal 2018; 44: 158–70.
- 32Chan CY, Huang SY, Sheu JJ, et al. Transcription factor HBP1 is a direct anti-cancer target of transcription factor FOXO1 in invasive oral cancer. Oncotarget 2017; 8: 14537–48.
- 33Dufait I, Van Valckenborgh E, Menu E, et al. Signal transducer and activator of transcription 3 in myeloid-derived suppressor cells: an opportunity for cancer therapy. Oncotarget 2016; 7: 42698–715.
- 34Fu F, Jiang W, Zhou L, et al. Circulating Exosomal miR-17-5p and miR-92a-3p predict pathologic stage and grade of colorectal cancer. Transl Oncol 2018; 11: 221–32.
- 35Dinh TK, Fendler W, Chalubinska-Fendler J, et al. Circulating miR-29a and miR-150 correlate with delivered dose during thoracic radiation therapy for non-small cell lung cancer. Radiat Oncol 2016; 11: 61.
- 36Sun Z, Wang S, Zhao RC. The roles of mesenchymal stem cells in tumor inflammatory microenvironment. J Hematol Oncol 2014;7:14.
- 37Shahar T, Rozovski U, Hess KR, et al. Percentage of mesenchymal stem cells in high-grade glioma tumor samples correlates with patient survival. Neuro Oncol 2017; 19: 660–8.
- 38Martinez VG, Ontoria-Oviedo I, Ricardo CP, et al. Overexpression of hypoxia-inducible factor 1 alpha improves immunomodulation by dental mesenchymal stem cells. Stem Cell Res Ther 2017;8:208.
- 39Whiteside TL. Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment. Semin Immunol 2018; 35: 69–79.