Negative regulation of tumor-infiltrating NK cell in clear cell renal cell carcinoma patients through the exosomal pathway

Yang Xia, Qiongfang Zhang, Quan Zhen, Yan Zhao, Nanjing Liu, Ting Li, Yanni Hao, Yao Zhang, Chunli Luo, Xiaohou Wu, Yang Xia, Qiongfang Zhang, Quan Zhen, Yan Zhao, Nanjing Liu, Ting Li, Yanni Hao, Yao Zhang, Chunli Luo, Xiaohou Wu

Abstract

Natural killer cells are the key components in tumor immunity and defects in function are necessary for tumor immune escape. Emerging studies on tumor cell-derived exosomes have shown the biological significance in tumor microenvironment, but the underlying role of exosomes in regulating natural killer cells functions in clear cell renal cell carcinoma patients remains unknown. Firstly, we precisely characterized the phenotype and function of natural killer cells in clear cell renal cell carcinoma patients vs healthy controls. With an inhibitory phenotype, tumor-infiltrating natural killer cells exhibited poor cytotoxic capacity and deficient potential to produce cytokines compared with natural killer cells from tumor margin tissue and non-tumor tissue. Next, we revealed that primary tumor cells trigged natural killer cell dysfunction in an exosome-dependent manner. Interestingly, exosomes from primary tumor cells were preferentially enriched with TGF-β1 which acted as important mediator of natural killer cell functional deficiency. In vitro culture of exosomes induced natural killer cell dysfunction mediated by activation of the TGF-β/SMAD signaling pathway, and abrogated by knockdown TGF-β. Our data indicate that exosomes from clear cell renal cell carcinoma induce natural killer cells dysfunction by regulating the TGF-β/SMAD pathway to evade innate immune surveillance.

Keywords: clear cell renal cell carcinoma; exosome; natural killer; transforming growth factor-β1; tumor immune evasion.

Conflict of interest statement

CONFLICTS OF INTEREST

All authors declare that they have no competing interests.

Figures

Figure 1. TINK cells in RCC display…
Figure 1. TINK cells in RCC display an abnormal phenotype
(A) Lymphocyte gate of freshly purified mononuclear cells (MNCs) and NK cells. (B) Percentage of NK cells from tumor tissue (T, n = 36), tumor margin tissue (M, n = 36), and non-tumor tissue (NT, n = 16, mean±sd). (C) Percentage of NK cells in PBMCs from ccRCC patients (ccRCC, n = 36) and healthy individuals (HC, n = 36, mean±sd). (D) The percentage of NK-cell activation receptors NKp46, NKG2D, NKG2C and inhibitory receptors NKG2A, CD158a, CD158b expressed on NK cells from tumor tissue (T, n = 36), tumor margin tissue (M, n = 36), and non-tumor tissue (NT, n = 16, mean±sd). (E) Pooled percentage of NK receptors expressed on NK cells from tumor tissue, tumor margin tissue, and non-tumor tissue (T, n = 36; M, n = 36; NT, n = 16, mean±sd), *p < 0.05, **p < 0.01 and ***p < 0.001vs corresponding control.
Figure 2. TINK cells in RCC exhibit…
Figure 2. TINK cells in RCC exhibit a functional deficiency and are associated with tumor progression
(A) The percentage of CD107a, IFN-γ and TNF-α expressed on NK cells from tumor tissue (T, n = 36), tumor margin tissue (M, n = 36), and non-tumor tissue (NT, n = 16, mean±sd). (B) The cytolytic activity of NK cells from different tissues cocultured with K562. (C) NK functional markers (CD107a, IFN-γ and TNF-α) expressed on TINK cells in patients with tumor diameters ≥7 cm (n = 13) VS patients with tumor diameters < 7 cm (n =23). (D) The percentage of CD107a, IFN-γ and TNF-α expressed on TINK cells decreases as tumors progress (n = 36). *p < 0.05, **p < 0.01 and ***p < 0.001vs corresponding control.
Figure 3. NK cell function was impaired…
Figure 3. NK cell function was impaired via exosome
(A) Transwell coculture model was used for the co-culture of primary NK cells and ccRCC cells. (B) Representative micrographs of immunofluorescence staining of cytokeratin and CAIX. DAPI counterstains nuclei in blue. 200× magnification. (C, D) NK cell functions (CD107a degranulation, IFN-γ and TNF-α production) in NT, M, T+GW4869, T+transwell and T groups. (E) Cytotoxicity of NK cells coculture with NT, M, T+GW4869, T+transwell, T and anti-TGFβ1 groups. All data shown were from three independent experiments. Error bars indicate ±SD, *p < 0.05, **p < 0.01 and ***p < 0.001vs corresponding control.
Figure 4. Exosome characterization
Figure 4. Exosome characterization
(A) Transmission Electron Microscopy (TEM). (B) Western Blot analysis for exosomal marker in exosomes and cell lysate samples. (C) The quantification and size distribution analysis of exosomes. Size distribution of exosomes derived from tumor and non-tumor tissues were measured by qNano system. Bar Chart showing the mean diameter (D) and concentration for exosomes from NT and T (E). Statistical analysis of the correlations between exosomal protein concentrations and clinical parameters, including tumor size (F) and TNM stage (G). All data shown were from three independent experiments. Error bars indicate ±SD, *p < 0.05, **p < 0.01 and ***p < 0.001vs corresponding control.
Figure 5. Texs treatment suppresses the cytolytic…
Figure 5. Texs treatment suppresses the cytolytic activity of NK cells in vitro
(A) Cytotoxicity of NK cells coculture with Texs at different concentrations. (B, C) The level of CD107a, IFN-γ and TNF-α production of NK cells in coculture with Texs from different stages. (D) Cytotoxicity of NK cells coculture with Texs from different stages. All data shown were from three independent experiments. Error bars indicate ±SD, *p < 0.05, **p < 0.01 and ***p < 0.001vs corresponding control.
Figure 6. Texs interacted with NK cells…
Figure 6. Texs interacted with NK cells through TGFβ1/Smad signaling
(A) TGFβ1 were measured by ELISA in NT-exos, T-exos, M-exos, 786-O exos and HK-2 exos. High expressions of TGFβ1 protein were found in T-exos and 786-O. (B) Western blot of TGF-β1 protein. (C) Exos and NK cells were cocultured together to access the cytotoxicity of NK cells. Exos could impair the cytotoxicity of NK cell, and TGF-β1 knockdown could reverse it. (D) Knockdown of TGF-β1 could inhibit the smad signaling expression. All data shown were from three independent experiments. Error bars indicate ±SD, *p < 0.05, **p < 0.01 and ***p < 0.001vs corresponding control.
Figure 7. A schematic illustration showed that…
Figure 7. A schematic illustration showed that clear cell renal carcinoma cell-derived exosomes could induce NK cells dysfunction by activating Smad signaling through exosomal surface TGF-β1

References

    1. Ljungberg B, Campbell SC, Choi HY, Jacqmin D, Lee JE, Weikert S, Kiemeney LA. The epidemiology of renal cell carcinoma. European urology. 2011;60:615–621.
    1. Rini BI, Campbell SC, Escudier B. Renal cell carcinoma. Lancet. 2009;373:1119–1132.
    1. Whiteside TL. Immune suppression in cancer: effects on immune cells, mechanisms and future therapeutic intervention. Seminars in cancer biology. 2006;16:3–15.
    1. Webster WS, Lohse CM, Thompson RH, Dong H, Frigola X, Dicks DL, Sengupta S, Frank I, Leibovich BC, Blute ML, Cheville JC, Kwon ED. Mononuclear cell infiltration in clear-cell renal cell carcinoma independently predicts patient survival. Cancer. 2006;107:46–53.
    1. Moretta L, Bottino C, Pende D, Mingari MC, Biassoni R, Moretta A. Human natural killer cells: their origin, receptors and function. European journal of immunology. 2002;32:1205–1211.
    1. Guillerey C, Huntington ND, Smyth MJ. Targeting natural killer cells in cancer immunotherapy. Nature immunology. 2016;17:1025–1036.
    1. Smyth MJ, Hayakawa Y, Takeda K, Yagita H. New aspects of natural-killer-cell surveillance and therapy of cancer. Nature reviews Cancer. 2002;2:850–861.
    1. Wu J, Lanier LL. Natural killer cells and cancer. Advances in cancer research. 2003;90:127–156.
    1. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, McKenna D, TE Le C Defor, Burns LJ, Orchard PJ, Blazar BR, Wagner JE, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105:3051–3057.
    1. Dutta A, Banerjee A, Saikia N, Phookan J, Baruah MN, Baruah S. Negative regulation of natural killer cell in tumor tissue and peripheral blood of oral squamous cell carcinoma. Cytokine. 2015;76:123–130.
    1. Frankenberger B, Noessner E, Schendel DJ. Immune suppression in renal cell carcinoma. Seminars in cancer biology. 2007;17:330–343.
    1. Bang C, Thum T. Exosomes: new players in cell-cell communication. The international journal of biochemistry & cell biology. 2012;44:2060–2064.
    1. Hazan-Halevy I, Rosenblum D, Weinstein S, Bairey O, Raanani P, Peer D. Cell-specific uptake of mantle cell lymphoma-derived exosomes by malignant and non-malignant B-lymphocytes. Cancer letters. 2015;364:59–69.
    1. Camussi G, Deregibus MC, Bruno S, Grange C, Fonsato V, Tetta C. Exosome/microvesicle-mediated epigenetic reprogramming of cells. American journal of cancer research. 2011;1:98–110.
    1. Wang W, Lotze MT. Good things come in small packages: exosomes, immunity and cancer. Cancer gene therapy. 2014;21:139–141.
    1. Pogge von Strandmann E, Simhadri VR, von Tresckow B, Sasse S, Reiners KS, Hansen HP, Rothe A, Boll B, Simhadri VL, Borchmann P, McKinnon PJ, Hallek M, Engert A. Human leukocyte antigen-B-associated transcript 3 is released from tumor cells and engages the NKp30 receptor on natural killer cells. Immunity. 2007;27:965–974.
    1. Lv LH, Wan YL, Lin Y, Zhang W, Yang M, Li GL, Lin HM, Shang CZ, Chen YJ, Min J. Anticancer drugs cause release of exosomes with heat shock proteins from human hepatocellular carcinoma cells that elicit effective natural killer cell antitumor responses in vitro. The Journal of biological chemistry. 2012;287:15874–15885.
    1. Liu C, Yu S, Zinn K, Wang J, Zhang L, Jia Y, Kappes JC, Barnes S, Kimberly RP, Grizzle WE, Zhang HG. Murine mammary carcinoma exosomes promote tumor growth by suppression of NK cell function. Journal of immunology. 2006;176:1375–1385.
    1. Szczepanski MJ, Szajnik M, Welsh A, Whiteside TL, Boyiadzis M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-beta1. Haematologica. 2011;96:1302–1309.
    1. Yamada N, Tsujimura N, Kumazaki M, Shinohara H, Taniguchi K, Nakagawa Y, Naoe T, Akao Y. Colorectal cancer cell-derived microvesicles containing microRNA-1246 promote angiogenesis by activating Smad 1/5/8 signaling elicited by PML down-regulation in endothelial cells. Biochimica et biophysica acta. 2014;1839:1256–1272.
    1. Shabtai M, Ye H, Frischer Z, Martin J, Waltzer WC, Malinowski K. Increased expression of activation markers in renal cell carcinoma infiltrating lymphocytes. The Journal of urology. 2002;168:2216–2219.
    1. Eckl J, Buchner A, Prinz PU, Riesenberg R, Siegert SI, Kammerer R, Nelson PJ, Noessner E. Transcript signature predicts tissue NK cell content and defines renal cell carcinoma subgroups independent of TNM staging. J Mol Med (Berl) 2012;90:55–66.
    1. Giraldo NA, Becht E, Remark R, Damotte D, Sautes-Fridman C, Fridman WH. The immune contexture of primary and metastatic human tumours. Current opinion in immunology. 2014;27:8–15.
    1. Liu Y, Zhao L, Li D, Yin Y, Zhang CY, Li J, Zhang Y. Microvesicle-delivery miR-150 promotes tumorigenesis by up-regulating VEGF, and the neutralization of miR-150 attenuate tumor development. Protein & cell. 2013;4:932–941.
    1. Muturi HT, Dreesen JD, Nilewski E, Jastrow H, Giebel B, Ergun S, Singer BB. Tumor and endothelial cell-derived microvesicles carry distinct CEACAMs and influence T-cell behavior. PloS one. 2013;8:e74654.
    1. Vitale M, Cantoni C, Pietra G, Mingari MC, Moretta L. Effect of tumor cells and tumor microenvironment on NK-cell function. European journal of immunology. 2014;44:1582–1592.
    1. Castriconi R, Cantoni C, Della Chiesa M, Vitale M, Marcenaro E, Conte R, Biassoni R, Bottino C, Moretta L, Moretta A. Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells. Proceedings of the National Academy of Sciences of the United States of America; 2003. pp. 4120–4125.
    1. Lee JC, Lee KM, Kim DW, Heo DS. Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. Journal of immunology. 2004;172:7335–7340.
    1. Jurisic V, Spuzic I, Konjevic G. A comparison of the NK cell cytotoxicity with effects of TNF-alpha against K-562 cells, determined by LDH release assay. Cancer letters. 1999;138:67–72.
    1. Pradier A, Passweg J, Villard J, Kindler V. Human bone marrow stromal cells and skin fibroblasts inhibit natural killer cell proliferation and cytotoxic activity. Cell transplantation. 2011;20:681–691.
    1. Thery C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Current protocols in cell biology. 2006 Chapter 3: Unit 3.22.
    1. Zhang HG, Hyde K, Page GP, Brand JP, Zhou J, Yu S, Allison DB, Hsu HC, Mountz JD. Novel tumor necrosis factor alpha-regulated genes in rheumatoid arthritis. Arthritis and rheumatism. 2004;50:420–431.
    1. de Vrij J, Maas SL, van Nispen M, Sena-Esteves M, Limpens RW, Koster AJ, Leenstra S, Lamfers ML, Broekman ML. Quantification of nanosized extracellular membrane vesicles with scanning ion occlusion sensing. Nanomedicine (Lond) 2013;8:1443–1458.

Source: PubMed

3
Tilaa