DOI QR코드

DOI QR Code

Acetate decreases PVR/CD155 expression via PI3K/AKT pathway in cancer cells

  • Tran, Na Ly (Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST)) ;
  • Lee, In Kyu (Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST)) ;
  • Choi, Jungkyun (Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST)) ;
  • Kim, Sang-Heon (Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST)) ;
  • Oh, Seung Ja (Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST))
  • Received : 2021.05.06
  • Accepted : 2021.07.05
  • Published : 2021.08.31

Abstract

In recent years, restoring anti-tumor immunity has garnered a growing interest in cancer treatment. As potential therapeutics, immune checkpoint inhibitors have demonstrated benefits in many clinical studies. Although various methods have been applied to suppress immune checkpoints to boost anti-tumor immunity, including the use of immune checkpoint inhibitors, there are still unmet clinical needs to improve the response rate of cancer treatment. Here, we show that acetate can suppress the expression of poliovirus receptor (PVR/CD155), a ligand for immune checkpoint, in colon cancer cells. We demonstrated that acetate treatment could enhance effector responses of CD8+ T cells by decreasing the expression of PVR/CD155 in cancer cells. We also found that acetate could reduce the expression of PVR/CD155 by deactivating the PI3K/AKT pathway. These results demonstrate that acetate-mediated expression of PVR/CD155 in cancer cells might potentiate the anti-tumor immunity in the microenvironment of cancer. Our findings indicate that maintaining particular acetate concentrations could be a complementary strategy in current cancer treatment.

Keywords

Acknowledgement

This work was supported by the Technology Innovation Program (10063334) funded by the Ministry of Trade, Industry & Energy. This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (2020R1C1C1009507) and the KIST Institutional Program to S.J.O.

References

  1. Siegel RL, Miller KD and Jemal A (2018) Cancer statistics, 2018. CA Cancer J Clin 68, 7-30 https://doi.org/10.3322/caac.21442
  2. Le DT, Hubbard-Lucey VM, Morse MA et al (2017) A blueprint to advance colorectal cancer immunotherapies. Cancer Immunol Res 5, 942-949 https://doi.org/10.1158/2326-6066.CIR-17-0375
  3. Le DT, Uram JN, Wang H et al (2015) PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 372, 2509-2520 https://doi.org/10.1056/NEJMoa1500596
  4. Cummings JH, Pomare EW, Branch WJ, Naylor CP and Macfarlane GT (1987) Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 28, 1221-1227 https://doi.org/10.1136/gut.28.10.1221
  5. Topping DL and Clifton PM (2001) Short-chain fatty acids and human colonic function: roles of resistant starch and nonstarch polysaccharides. Physiol Rev 81, 1031-1064 https://doi.org/10.1152/physrev.2001.81.3.1031
  6. Marques C, Oliveira CS, Alves S et al (2013) Acetateinduced apoptosis in colorectal carcinoma cells involves lysosomal membrane permeabilization and cathepsin D release. Cell Death Dis 4, e507 https://doi.org/10.1038/cddis.2013.29
  7. Lin Y, Ma CC, Liu CK et al (2016) NMR-based fecal metabolomics fingerprinting as predictors of earlier diagnosis in patients with colorectal cancer. Oncotarget 7, 29454-29464 https://doi.org/10.18632/oncotarget.8762
  8. Kucan Brlic P, Lenac Rovis T, Cinamon G, Tsukerman P, Mandelboim O and Jonjic S (2019) Targeting PVR (CD155) and its receptors in anti-tumor therapy. Cell Mol Immunol 16, 40-52 https://doi.org/10.1038/s41423-018-0168-y
  9. Alex S, Lange K, Amolo T et al (2013) Short-chain fatty acids stimulate angiopoietin-like 4 synthesis in human colon adenocarcinoma cells by activating peroxisome proliferatoractivated receptor gamma. Mol Cell Biol 33, 1303-1316 https://doi.org/10.1128/MCB.00858-12
  10. Shaib W, Mahajan R and El-Rayes B (2013) Markers of resistance to anti-EGFR therapy in colorectal cancer. J Gastrointest Oncol 4, 308-318 https://doi.org/10.3978/j.issn.2078-6891.2013.029
  11. Parada Venegas D, De la Fuente MK, Landskron G et al (2019) Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol 10, 277 https://doi.org/10.3389/fimmu.2019.00277
  12. Wang G, Huang S, Wang Y et al (2019) Bridging intestinal immunity and gut microbiota by metabolites. Cell Mol Life Sci 76, 3917-3937 https://doi.org/10.1007/s00018-019-03190-6
  13. Okumura G, Iguchi-Manaka A, Murata R, Yamashita-Kanemaru Y, Shibuya A and Shibuya K (2020) Tumor-derived soluble CD155 inhibits DNAM-1-mediated antitumor activity of natural killer cells. J Exp Med 217, 1
  14. Sanchez-Correa B, Valhondo I, Hassouneh F et al (2019) DNAM-1 and the TIGIT/PVRIG/TACTILE axis: novel immune checkpoints for natural killer cell-based cancer immunotherapy. Cancers (Basel) 11, 877 https://doi.org/10.3390/cancers11060877
  15. Vlahos CJ, Matter WF, Hui KY and Brown RF (1994) A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002). J Biol Chem 269, 5241-5248 https://doi.org/10.1016/S0021-9258(17)37680-9
  16. Kamran N, Takai Y, Miyoshi J, Biswas SK, Wong JS and Gasser S (2013) Toll-like receptor ligands induce expression of the costimulatory molecule CD155 on antigen-presenting cells. PLoS One 8, e54406 https://doi.org/10.1371/journal.pone.0054406
  17. Sung B, Pandey MK, Ahn KS et al (2008) Anacardic acid (6-nonadecyl salicylic acid), an inhibitor of histone acetyltransferase, suppresses expression of nuclear factor-kappaBregulated gene products involved in cell survival, proliferation, invasion, and inflammation through inhibition of the inhibitory subunit of nuclear factor-kappaBalpha kinase, leading to potentiation of apoptosis. Blood 111, 4880-4891 https://doi.org/10.1182/blood-2007-10-117994
  18. Zheng Q, Wang B, Gao J et al (2018) CD155 knockdown promotes apoptosis via AKT/Bcl-2/Bax in colon cancer cells. J Cell Mol Med 22, 131-140 https://doi.org/10.1111/jcmm.13301
  19. Enloe BM and Jay DG (2011) Inhibition of Necl-5 (CD155/PVR) reduces glioblastoma dispersal and decreases MMP-2 expression and activity. J Neurooncol 102, 225-235 https://doi.org/10.1007/s11060-010-0323-5
  20. Weiner GJ (2015) Building better monoclonal antibody-based therapeutics. Nat Rev Cancer 15, 361-370 https://doi.org/10.1038/nrc3930
  21. Marin-Acevedo JA, Dholaria B, Soyano AE, Knutson KL, Chumsri S and Lou Y (2018) Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol 11, 39 https://doi.org/10.1186/s13045-018-0582-8
  22. Pardoll DM (2012) The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12, 252-264 https://doi.org/10.1038/nrc3239
  23. Egelston CA, Avalos C, Tu TY et al (2018) Human breast tumor-infiltrating CD8(+) T cells retain polyfunctionality despite PD-1 expression. Nat Commun 9, 4297 https://doi.org/10.1038/s41467-018-06653-9
  24. Nelson N, Lopez-Pelaez M, Palazon A et al (2019) A cell-engineered system to assess tumor cell sensitivity to CD8(+) T cell-mediated cytotoxicity. Oncoimmunology 8, 1599635
  25. Wei J, Long L, Zheng W et al (2019) Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy. Nature 576, 471-476 https://doi.org/10.1038/s41586-019-1821-z