2,687
Views
15
CrossRef citations to date
0
Altmetric
Research Paper

DNA methylation in the promoters of PD-L1, MMP9, ARG1, galectin-9, TIM-3, VISTA and TGF-β genes in HLA-DR myeloid cells, compared with HLA-DR+ antigen-presenting cells

, , , ORCID Icon, ORCID Icon & ORCID Icon
Pages 1275-1288 | Received 05 Mar 2020, Accepted 01 May 2020, Published online: 18 May 2020

References

  • Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162–174.
  • Ennaciri J, Girard D. Immune system: maturation of myeloid cells. Methods Mol Biol. 2009;550:195–203.
  • Awad RM, De Vlaeminck Y, Maebe J, et al. Turn back the time: targeting tumor infiltrating myeloid cells to revert cancer progression. Front Immunol. 2018;9:1977.
  • Cripps JG, Gorham JD. MDSC in autoimmunity. Int Immunopharmacol. 2011;11(7):789–793.
  • Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor cells in cancer: recent progress and prospects. Immunol Cell Biol. 2013;91(8):493–502.
  • O’Connor MA, Rastad JL, Green WR. The role of myeloid-derived suppressor cells in viral infection. Viral Immunol. 2017;30(2):82–97.
  • Yang T, Li J, Li R, et al. Correlation between MDSC and immune tolerance in transplantation: cytokines, pathways and cell-cell interaction. Curr Gene Ther. 2019;19(2):81–92.
  • Makarenkova VP, Bansal V, Matta BM, et al. CD11b + /Gr-1 + myeloid suppressor cells cause T cell dysfunction after traumatic stress. J Immunol. 2006;176(4):2085–2094.
  • Lindau D, Gielen P, Kroesen M, et al. The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology. 2013;138(2):105–115.
  • Schlecker E, Stojanovic A, Eisen C, et al. Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J Immunol. 2012;189(12):5602–5611.
  • Bernal-Estevez DA, Garcia O, Sanchez R, et al. Monitoring the responsiveness of T and antigen presenting cell compartments in breast cancer patients is useful to predict clinical tumor response to neoadjuvant chemotherapy. BMC Cancer. 2018;18(1):77.
  • Weber R, Fleming V, Hu X, et al. Myeloid-derived suppressor cells hinder the anti-cancer activity of immune checkpoint inhibitors. Front Immunol. 2018;9:1310.
  • Platten M, Wick W, Van den Eynde BJ. Tryptophan catabolism in cancer: beyond IDO and tryptophan depletion. Cancer Res. 2012;72(21):5435–5440.
  • Meier A, Bagchi A, Sidhu HK, et al. Upregulation of PD-L1 on monocytes and dendritic cells by HIV-1 derived TLR ligands. AIDS. 2008;22(5):655–658.
  • Anderson AC, Anderson DE, Bregoli L, et al. Promotion of tissue inflammation by the immune receptor Tim-3 expressed on innate immune cells. Science. 2007;318(5853):1141–1143.
  • Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunol Rev. 2017;276(1):97–111.
  • Gao J, Ward JF, Pettaway CA, et al. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med. 2017;23(5):551–555.
  • Limagne E, Richard C, Thibaudin M, et al. Tim-3/galectin-9 pathway and mMDSC control primary and secondary resistances to PD-1 blockade in lung cancer patients. Oncoimmunology. 2019;8(4):e1564505.
  • Goncalves Silva I, Yasinska IM, Sakhnevych SS, et al. The Tim-3-galectin-9 secretory pathway is involved in the immune escape of human acute myeloid leukemia cells. EBioMedicine. 2017;22:44–57.
  • Dardalhon V, Anderson AC, Karman J, et al. Tim-3/galectin-9 pathway: regulation of Th1 immunity through promotion of CD11b+Ly-6G+ myeloid cells. J Immunol. 2010;185(3):1383–1392.
  • Deng J, Li J, Sarde A, et al. Hypoxia-induced VISTA promotes the suppressive function of myeloid-derived suppressor cells in the tumor microenvironment. Cancer Immunol Res. 2019;7(7):1079–1090.
  • Wang L, Jia B, Claxton DF, et al. VISTA is highly expressed on MDSCs and mediates an inhibition of T cell response in patients with AML. Oncoimmunology. 2018;7(9):e1469594.
  • Wang L, Rubinstein R, Lines JL, et al. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J Exp Med. 2011;208(3):577–592.
  • Crook KR. Role of myeloid-derived suppressor cells in autoimmune disease. World J Immunol. 2014;4(1):26–33.
  • Li M, Zhu D, Wang T, et al. Roles of myeloid-derived suppressor cell subpopulations in autoimmune arthritis. Front Immunol. 2018;9:2849.
  • Putiri EL, Robertson KD. Epigenetic mechanisms and genome stability. Clin Epigenetics. 2011;2(2):299–314.
  • Mazzio EA, Soliman KF. Basic concepts of epigenetics: impact of environmental signals on gene expression. Epigenetics. 2012;7(2):119–130.
  • Zhang C, Wang S, Liu Y, et al. Epigenetics in myeloid derived suppressor cells: a sheathed sword towards cancer. Oncotarget. 2016;7(35):57452–57463.
  • Lu Q. The critical importance of epigenetics in autoimmunity. J Autoimmun. 2013;41:1–5.
  • Suarez-Alvarez B, Baragano Raneros A, Ortega F, et al. Epigenetic modulation of the immune function: a potential target for tolerance. Epigenetics. 2013;8(7):694–702.
  • Sasidharan Nair V, Saleh R, Toor SM, et al. Transcriptomic profiling disclosed the role of DNA methylation and histone modifications in tumor-infiltrating myeloid-derived suppressor cell subsets in colorectal cancer. Clin Epigenetics. 2020;12(1):13.
  • Toor SM, Syed Khaja AS, El Salhat H, et al. Myeloid cells in circulation and tumor microenvironment of breast cancer patients. Cancer Immunol Immunother. 2017;66(6):753–764.
  • Toor SM, Elkord E. Therapeutic prospects of targeting myeloid-derived suppressor cells and immune checkpoints in cancer. Immunol Cell Biol. 2018;96(9):888–897.
  • Esteller M. CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future. Oncogene. 2002;21(35):5427–5440.
  • Sun B, Hu L, Luo Z-Y, et al. DNA methylation perspectives in the pathogenesis of autoimmune diseases. Clin Immunol. 2016;164:21–27.
  • Pastor WA, Aravind L, Rao A. TETonic shift: biological roles of TET proteins in DNA demethylation and transcription. Nat Rev Mol Cell Biol. 2013;14(6):341–356.
  • Huang Y, Rao A. Connections between TET proteins and aberrant DNA modification in cancer. Trends Genet. 2014;30(10):464–474.
  • Elashi AA, Sasidharan Nair V, Taha RZ, et al. DNA methylation of immune checkpoints in the peripheral blood of breast and colorectal cancer patients. Oncoimmunology. 2019;8(2):e1542918.
  • Sasidharan Nair V, El Salhat H, Taha RZ, et al. DNA methylation and repressive H3K9 and H3K27 trimethylation in the promoter regions of PD-1, CTLA-4, TIM-3, LAG-3, TIGIT, and PD-L1 genes in human primary breast cancer. Clin Epigenetics. 2018;10(1):78.
  • Nair VS, Song MH, Ko M, et al. DNA demethylation of the Foxp3 enhancer is maintained through modulation of ten-eleven-translocation and DNA methyltransferases. Mol Cells. 2016;39(12):888–897.
  • Maier T, Guell M, Serrano L. Correlation of mRNA and protein in complex biological samples. FEBS Lett. 2009;583(24):3966–3973.
  • Turek-Plewa J, Jagodzinski PP. The role of mammalian DNA methyltransferases in the regulation of gene expression. Cell Mol Biol Lett. 2005;10(4):631–647.
  • Bestor TH. Activation of mammalian DNA methyltransferase by cleavage of a Zn binding regulatory domain. Embo J. 1992;11(7):2611–2617.
  • Hopfer OJ, Komor M, Koehler IS, et al. Expression of DNMT isoforms is differentially associated with aberrant promotor methylation in MDS hematopoietic progenitor cells during lineage specific differentiation. Blood. 2006;108(11):2628.
  • Li S, Chiang T-C, Richard-Davis G, et al. DNA hypomethylation and imbalanced expression of DNA methyltransferases (DNMT1, 3A, and 3B) in human uterine leiomyoma. Gynecol Oncol. 2003;90(1):123–130.
  • Gustafsson JR, Katsioudi G, Degn M, et al. DNMT1 regulates expression of MHC class I in post-mitotic neurons. Mol Brain. 2018;11(1):36.
  • Zhong W, Li B, Xu Y, et al. Hypermethylation of the micro-RNA 145 promoter is the key regulator for NLRP3 inflammasome-induced activation and plaque formation. JACC Basic Transl Sci. 2018;3(5):604–624.
  • Mishra M, Kowluru RA. Epigenetic modification of mitochondrial DNA in the development of diabetic retinopathy. Invest Ophthalmol Vis Sci. 2015;56(9):5133–5142.
  • Lee C-R, Kwak Y, Yang T, et al. Myeloid-derived suppressor cells are controlled by regulatory T cells via TGF-β during murine colitis. Cell Rep. 2016;17(12):3219–3232.
  • Aoki CA, Borchers AT, Li M, et al. Transforming growth factor beta (TGF-beta) and autoimmunity. Autoimmun Rev. 2005;4(7):450–459.
  • Sun C, Mezzadra R, Schumacher TN. Regulation and function of the PD-L1 checkpoint. Immunity. 2018;48(3):434–452.
  • Sato R, Imamura K, Sakata S, et al. Disorder of coagulation-fibrinolysis system: an emerging toxicity of anti-PD-1/PD-L1 monoclonal antibodies. J Clin Med. 2019;8(6):6. .
  • Bogdan C. Nitric oxide and the immune response. Nat Immunol. 2001;2(10):907–916.
  • Nagaraj S, Gupta K, Pisarev V, et al. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med. 2007;13(7):828–835.
  • Budhwar S, Verma P, Verma R, et al. The Yin and Yang of myeloid derived suppressor cells. Front Immunol. 2018;9:2776.
  • Gonzalez-Junca A, Driscoll KE, Pellicciotta I, et al. Autocrine TGFβ is a survival factor for monocytes and drives immunosuppressive lineage commitment. Cancer Immunol Res. 2019;7(2):306–320.
  • Shvedova AA, Kisin ER, Yanamala N, et al. MDSC and TGFβ are required for facilitation of tumor growth in the lungs of mice exposed to carbon nanotubes. Cancer Res. 2015;75(8):1615–1623.
  • Saleh R, Elkord E. Acquired resistance to cancer immunotherapy: role of tumor-mediated immunosuppression. Semin Cancer Biol. 2019. DOI:10.1016/j.semcancer.2019.07.017
  • Sanjabi S, Oh SA, Li MO. Regulation of the immune response by TGF-β: from conception to autoimmunity and infection. Cold Spring Harb Perspect Biol. 2017;9(6):6.
  • Wegner A, Verhagen J, Wraith DC. Myeloid-derived suppressor cells mediate tolerance induction in autoimmune disease. Immunology. 2017;151(1):26–42.
  • Lee S-E, Lim J-Y, Kim TW, et al. Matrix metalloproteinase-9 in monocytic myeloid-derived suppressor cells correlate with early infections and clinical outcomes in allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2018;24(1):32–42.
  • Ram M, Sherer Y, Shoenfeld Y. Matrix metalloproteinase-9 and autoimmune diseases. J Clin Immunol. 2006;26(4):299–307.
  • Fleming V, Hu X, Weber R, et al. Targeting myeloid-derived suppressor cells to bypass tumor-induced immunosuppression. Front Immunol. 2018;9:398.
  • Mikyskova R, Indrova M, Vlkova V, et al. DNA demethylating agent 5-azacytidine inhibits myeloid-derived suppressor cells induced by tumor growth and cyclophosphamide treatment. J Leukoc Biol. 2014;95(5):743–753.
  • Terranova-Barberio M, Thomas S, Munster PN. Epigenetic modifiers in immunotherapy: a focus on checkpoint inhibitors. Immunotherapy. 2016;8(6):705–719.