DOI QR코드

DOI QR Code

Tumour-Derived Reg3A Educates Dendritic Cells to Promote Pancreatic Cancer Progression

  • Guo, Jie (Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology) ;
  • Liao, Mengfan (Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology) ;
  • Hu, Xianmin (Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology) ;
  • Wang, Jun (Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology)
  • Received : 2021.05.27
  • Accepted : 2021.07.22
  • Published : 2021.09.30

Abstract

As a pancreatic inflammatory marker, regenerating islet-derived protein 3A (Reg3A) plays a key role in inflammation-associated pancreatic carcinogenesis by promoting cell proliferation, inhibiting apoptosis, and regulating cancer cell migration and invasion. This study aimed to reveal a novel immuno-regulatory mechanism by which Reg3A modulates tumour-promoting responses during pancreatic cancer (PC) progression. In an in vitro Transwell system that allowed the direct co-culture of human peripheral blood-derived dendritic cells (DCs) and Reg3A-overexpressing/ silenced human PC cells, PC cell-derived Reg3A was found to downregulate CD80, CD83 and CD86 expression on educated DCs, increase DC endocytic function, inhibit DC-induced T lymphocyte proliferation, reduce IL-12p70 production, and enhance IL-23 production by DCs. The positive effect of tumour-derived Reg3A-educated human DCs on PC progression was demonstrated in vivo by intraperitoneally transferring them into PC-implanted severe combined immunodeficiency (SCID) mice reconstituted with human T cells. A Reg3A-JAK2/STAT3 positive feedback loop was identified in DCs educated with Reg3A. In conclusion, as a tumour-derived factor, Reg3A acted to block the differentiation and maturation of the most important antigen-presenting cells, DCs, causing them to limit their potential anti-tumour responses, thus facilitating PC escape and progression.

Keywords

Acknowledgement

This work was funded by grants from the National Natural Science Foundation of China (81602108, 71974153) and the Provincial College Students' Innovative Entrepreneurial Training Program in Hubei (201810488115).

References

  1. Balachandran, V.P., Beatty, G.L., and Dougan, S.K. (2019). Broadening the impact of immunotherapy to pancreatic cancer: challenges and opportunities. Gastroenterology 156, 2056-2072. https://doi.org/10.1053/j.gastro.2018.12.038
  2. Bazzichetto, C., Conciatori, F., Luchini, C., Simionato, F., Santoro, R., Vaccaro, V., Corbo, V., Falcone, I., Ferretti, G., Cognetti, F., et al. (2020). From genetic alterations to tumor microenvironment: the Ariadne's string in pancreatic cancer. Cells 9, 309. https://doi.org/10.3390/cells9020309
  3. Chen, G., Le, Y., Zhou, L., Gong, L., Li, X., Li, Y., Liao, Q., Duan, K., Tong, J., and Ouyang, W. (2016). Dexmedetomidine inhibits maturation and function of human cord blood-derived dendritic cells by interfering with synthesis and secretion of IL-12 and IL-23. PLoS One 11, e0153288. https://doi.org/10.1371/journal.pone.0153288
  4. Chen, I.L., Todd, I., Tighe, P.J., and Fairclough, L.C. (2020). Electronic cigarette vapour moderately stimulates pro-inflammatory signalling pathways and interleukin-6 production by human monocyte-derived dendritic cells. Arch. Toxicol. 94, 2097-2112. https://doi.org/10.1007/s00204-020-02757-8
  5. Chyuan, I.T. and Lai, J.H. (2020). New insights into the IL-12 and IL-23: from a molecular basis to clinical application in immune-mediated inflammation and cancers. Biochem. Pharmacol. 175, 113928. https://doi.org/10.1016/j.bcp.2020.113928
  6. Conejo-Garcia, J.R., Rutkowski, M.R., and Cubillos-Ruiz, J.R. (2016). State-of-the-art of regulatory dendritic cells in cancer. Pharmacol. Ther. 164, 97-104. https://doi.org/10.1016/j.pharmthera.2016.04.003
  7. Deicher, A., Andersson, R., Tingstedt, B., Lindell, G., Bauden, M., and Ansari, D. (2018). Targeting dendritic cells in pancreatic ductal adenocarcinoma. Cancer Cell Int. 18, 85. https://doi.org/10.1186/s12935-018-0585-0
  8. Dudek, A.M., Martin, S., Garg, A.D., and Agostinis, P. (2013). Immature, semi-mature, and fully mature dendritic cells: toward a DC-cancer cells interface that augments anticancer immunity. Front. Immunol. 4, 438. https://doi.org/10.3389/fimmu.2013.00438
  9. Foley, K., Kim, V., Jaffee, E., and Zheng, L. (2016). Current progress in immunotherapy for pancreatic cancer. Cancer Lett. 381, 244-251. https://doi.org/10.1016/j.canlet.2015.12.020
  10. Fukunaga, A., Miyamoto, M., Cho, Y., Murakami, S., Kawarada, Y., Oshikiri, T., Kato, K., Kurokawa, T., Suzuoki, M., Nakakubo, Y., et al. (2004). Cd8+ tumor-infiltrating lymphocytes together with Cd4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas 28, e26-e31. https://doi.org/10.1097/00006676-200401000-00023
  11. Gabrilovich, D. (2004). Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nat. Rev. Immunol. 4, 941-952. https://doi.org/10.1038/nri1498
  12. Giovanelli, P., Sandoval, T.A., and Cubillos-Ruiz, J.R. (2019). Dendritic cell metabolism and function in tumors. Trends Immunol. 40, 699-718. https://doi.org/10.1016/j.it.2019.06.004
  13. He, S., Fei, M., Wu, Y., Zheng, D., Wan, D., Wang, L., and Li, D. (2011). Distribution and clinical significance of Th17 cells in the tumor microenvironment and peripheral blood of pancreatic cancer patients. Int. J. Mol. Sci. 12, 7424-7437. https://doi.org/10.3390/ijms12117424
  14. Hegde, S., Krisnawan, V.E., Herzog, B.H., Zuo, C., Breden, M.A., Knolhoff, B.L., Hogg, G.D., Tang, J.P., Baer, J.M., Mpoy, C., et al. (2020). Dendritic cell paucity leads to dysfunctional immune surveillance in pancreatic cancer. Cancer Cell 37, 289-307.e9. https://doi.org/10.1016/j.ccell.2020.02.008
  15. Hirooka, S., Yanagimoto, H., Satoi, S., Yamamoto, T., Toyokawa, H., Yamaki, S., Yui, R., Inoue, K., Michiura, T., and Kwon, A.H. (2011). The role of circulating dendritic cells in patients with unresectable pancreatic cancer. Anticancer Res. 31, 3827-3834.
  16. Liu, X., Wang, J., Wang, H., Yin, G., Liu, Y., Lei, X., and Xiang, M. (2015). REG3A accelerates pancreatic cancer cell growth under IL-6-associated inflammatory condition: involvement of a REG3A-JAK2/STAT3 positive feedback loop. Cancer Lett. 362, 45-60. https://doi.org/10.1016/j.canlet.2015.03.014
  17. Liu, X., Zhou, Z., Cheng, Q., Wang, H., Cao, H., Xu, Q., Tuo, Y., Jiang, L., Zou, Y., Ren, H., et al. (2017). Acceleration of pancreatic tumorigenesis under immunosuppressive microenvironment induced by Reg3g overexpression. Cell Death Dis. 8, e3033. https://doi.org/10.1038/cddis.2017.424
  18. Ma, C., Su, M., Shen, K., Chen, J., Ning, Y., and Qi, C. (2020). Key genes and pathways in tumor-educated dendritic cells by bioinformatical analysis. Microbiol. Immunol. 64, 63-71. https://doi.org/10.1111/1348-0421.12747
  19. Nefedova, Y., Cheng, P., Gilkes, D., Blaskovich, M., Beg, A.A., Sebti, S.M., and Gabrilovich, D.I. (2005). Activation of dendritic cells via inhibition of Jak2/STAT3 signaling. J. Immunol. 175, 4338-4346. https://doi.org/10.4049/jimmunol.175.7.4338
  20. Padoan, A., Plebani, M., and Basso, D. (2019). Inflammation and pancreatic cancer: focus on metabolism, cytokines, and immunity. Int. J. Mol. Sci. 20, 676. https://doi.org/10.3390/ijms20030676
  21. Ren, B., Cui, M., Yang, G., Wang, H., Feng, M., You, L., and Zhao, Y. (2018). Tumor microenvironment participates in metastasis of pancreatic cancer. Mol. Cancer 17, 108. https://doi.org/10.1186/s12943-018-0858-1
  22. Takasawa, S. (2016). Regenerating gene (REG) product and its potential clinical usage. Expert Opin. Ther. Targets 20, 541-550. https://doi.org/10.1517/14728222.2016.1123691
  23. Tjomsland, V., Spangeus, A., Sandstrom, P., Borch, K., Messmer, D., and Larsson, M. (2010). Semi mature blood dendritic cells exist in patients with ductal pancreatic adenocarcinoma owing to inflammatory factors released from the tumor. PLoS One 5, e13441. https://doi.org/10.1371/journal.pone.0013441
  24. Wang, J., Zhou, H., Han, Y., Liu, X., Wang, M., Wang, X., Yin, G., Li, X., and Xiang, M. (2014). SOCS3 methylation in synergy with Reg3A overexpression promotes cell growth in pancreatic cancer. J. Mol. Med. (Berl.) 92, 1257-1269. https://doi.org/10.1007/s00109-014-1184-8
  25. Yamamoto, T., Yanagimoto, H., Satoi, S., Toyokawa, H., Yamao, J., Kim, S., Terakawa, N., Takahashi, K., and Kwon, A.H. (2012). Circulating myeloid dendritic cells as prognostic factors in patients with pancreatic cancer who have undergone surgical resection. J. Surg. Res. 173, 299-308. https://doi.org/10.1016/j.jss.2010.09.027
  26. Yan, J., Smyth, M.J., and Teng, M.W.L. (2018). Interleukin (Il)-12 and Il-23 and their conflicting roles in cancer. Cold Spring Harb. Perspect. Biol. 10, a028530. https://doi.org/10.1101/cshperspect.a028530
  27. Yin, G., Du, J., Cao, H., Liu, X., Xu, Q., and Xiang, M. (2015). Reg3g promotes pancreatic carcinogenesis in a murine model of chronic pancreatitis. Dig. Dis. Sci. 60, 3656-3668. https://doi.org/10.1007/s10620-015-3787-5
  28. Zhang, M.Y., Wang, J., and Guo, J. (2019). Role of regenerating islet-derived protein 3A in gastrointestinal cancer. Front. Oncol. 9, 1449. https://doi.org/10.3389/fonc.2019.01449