Low-Dose Decitabine Inhibits Cytotoxic T Lymphocytes-Mediated Platelet Destruction via Modulating PD-1 Methylation in Immune Thrombocytopenia

Panpan Han, Tianshu Yu, Yu Hou, Yajing Zhao, Yang Liu, Yunqi Sun, Haoyi Wang, Pengcheng Xu, Guosheng Li, Tao Sun, Xiang Hu, Xinguang Liu, Lizhen Li, Jun Peng, Hai Zhou, Ming Hou, Panpan Han, Tianshu Yu, Yu Hou, Yajing Zhao, Yang Liu, Yunqi Sun, Haoyi Wang, Pengcheng Xu, Guosheng Li, Tao Sun, Xiang Hu, Xinguang Liu, Lizhen Li, Jun Peng, Hai Zhou, Ming Hou

Abstract

Cytotoxic T lymphocytes (CTLs)-mediated platelet destruction plays an important role in the pathogenesis of primary immune thrombocytopenia (ITP). The programmed cell death protein 1 (PD-1) signaling can turn off autoreactive T cells and induce peripheral tolerance. Herein, we found that the expression of PD-1 and its ligand PD-L1 on CD8+ T cells from ITP patients was decreased. Activating PD-1 pathway by PD-L1-Fc fusion protein inhibited CTLs-mediated platelet destruction in ITP in vitro. PD-1 promoter hypermethylation in CD8+ T cells was found in ITP patients, resulting in decreased PD-1 expression. The demethylating agent decitabine at a low dose was proved to restore the methylation level and expression of PD-1 on CD8+ T cells and reduce the cytotoxicity of CTLs of ITP patients. The phosphorylation levels of phosphatidylinositol 3-kinase (PI3K) and AKT in CD8+ T cells were significantly downregulated by low-dose decitabine. Furthermore, blocking PD-1 could counteract the effect of low-dose decitabine on CTLs from ITP patients. Therefore, our data suggest that the aberrant PD-1/PD-L1 pathway is involved in the pathophysiology of ITP and enhancing PD-1/PD-L1 signaling is a promising therapeutic approach for ITP management. Our results reveal the immunomodulatory mechanism of low-dose decitabine in ITP by inhibiting CTLs cytotoxicity to autologous platelets through PD-1 pathway.

Keywords: PD-1; PD-L1; cytotoxic T lymphocytes; decitabine; immune thrombocytopenia.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Han, Yu, Hou, Zhao, Liu, Sun, Wang, Xu, Li, Sun, Hu, Liu, Li, Peng, Zhou and Hou.

Figures

Figure 1
Figure 1
The expression of programed cell death protein 1 (PD-1) and PD-1 ligand (PD-L1) in immune thrombocytopenia (ITP) patients was lower than that of healthy controls. (A) The mRNA expression level of PD-1 in PBMCs from ITP patients had no difference with that of healthy controls (0.51 × 10−3 ± 0.39 × 10−3 vs. 0.37 × 10−3 ± 0.31 × 10−3, n = 13, P = 0.37, Unpaired t-test). (B,C) Significantly decreased PD-1 expression on CD8+ T cells (B, median; 2.70, IQR: 2.36–3.21, n = 35, vs. median: 2.56 IQR: 2.24–2.82, n = 40, P = 0.023, Mann Whitney test) and CD4+ T cells (C, 3.35 ± 0.53, n = 35 vs. 3.10 ± 0.45, n = 40, P = 0.032, Unpaired t-test) of ITP patients compared with healthy controls were observed. (D,E) Decreased PD-L1 expression on CD8+ T cells (D, 5.98 ± 1.35, n = 35 vs. 5.00 ± 1.77, n = 40, P = 0.010, Unpaired t-test) and CD4+ T cells (E, median: 5.58, IQR: 4.50–7.33, n = 35, vs. median: 2.25, IQR: 1.77–3.54, n = 40, P < 0.0001, Mann Whitney test) of ITP patients compared with healthy controls were observed. (F) The expression level of PD-L1 on platelet from ITP patients had no difference with that of healthy controls (median: 5.90, IQR: 5.42–6.31, n = 18, vs. median: 5.74, IQR: 5.34–6.61, n = 18, P = 0.894, Mann Whitney test). (G,H) Linear regression analysis of the PD-1 (G) and PD-L1 (H) expression on CD8+ T cells and platelet counts in ITP patients. *P < 0.05, **P < 0.01, ****P < 0.0001.
Figure 2
Figure 2
PD-L1-Fc reduced CTLs-mediated platelet apoptosis in ITP. (A) Platelets were gated by CD61 positive. (B) The gated dot-plots presented the apoptosis of platelets treated with PBS or PD-L1-Fc and cultured with CTLs of ITP patients treated with PBS or PD-L1-Fc. (C) A significant reduction was observed in CTLs-induced platelet apoptosis from ITP patients cultured with PD-L1-Fc in vitro after 72 h (9.76 ± 4.45 vs. 8.69 ± 4.29, n = 18, P = 0.006, Paired t-test). **P < 0.01.
Figure 3
Figure 3
Increased PD-1 methylation in ITP patients. (A) Bisulfite sequencing PCR analysis of PD-1 methylation. The 340-bp region (−975 to −636) containing 10 CpG residues in CpG rich area upstream of transcriptional start site (TSS) was amplified and sequenced. (B) The percentage of methylated residues in the PD-1 promoter of CD8+ T cells in ITP patients was significantly higher compared with healthy controls (position 1, 18.00 ± 14.83% vs. 58.00 ± 13.04%, P = 0.006; position 2, 30.00 ± 10.00% vs. 78.00 ± 13.04%, P = 0.002; position 3, 30.00 ± 10.00% vs. 72.00 ± 10.95%, P = 0.002; position 4, 34.00 ± 11.40% vs. 78.00 ± 13.04%, P = 0.002; position 5, 34.00 ± 16.73% vs. 74.00 ± 11.40%, P = 0.006; position 6, 18.00 ± 13.04% vs. 44.00 ± 15.17%, P = 0.02; position 7, 32.00 ± 8.37% vs. 66.00 ± 5.48%, P = 0.0006; position 8, 34.00 ± 5.48% vs. 70.00 ± 15.81%, P = 0.005; position 9, 34.00 ± 5.58% vs. 76.00 ± 13.42%, P = 0.002; position 10, 8.00 ± 4.47% vs. 30.00 ± 7.07%, P = 0.002, n = 5, Multiple t-tests). *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 4
Figure 4
Low-dose decitabine (Dec) increased PD-1 expression on CD8+ T cells and ameliorated thrombocytopenia in ITP. (A,B) Low-dose decitabine significantly increased PD-1 expression on CD8+ T cells in vitro (B, median: 2.33, IQR: 1.98–3.54 vs. median: 2.61, IQR: 2.09–4.18, n = 16, P = 0.001, Wilcoxon test). (C) Platelet counts in severe combined immunodeficiency (SCID) mice that received 5 × 104 splenocytes from either wild type C57 mice (♦, n = 10) treated with an equal volume of PBS, non-immunized CD61-knockout (KO) mice (▾, n = 10) treated with an equal volume of PBS, or platelet-immunized CD61-KO mice treated with an equal volume of PBS (•, n = 10) or decitabine (▴, n = 10). On day 21 and 28, a significantly higher platelet count was observed in the decitabine-treated group compared with PBS (day 21, 360.0 × 109 ± 27.04 × 109/L vs. 643.0 × 109 ± 45.97 × 109/L; P = 0.0005; day 28, 401.0 × 109 ± 23.87 × 109/L vs. 713.0 × 109 ± 52.35 × 109/L; P = 0.001). The horizontally dotted lines represent the normal mean platelet count ± SEM (dotted lines) from 40 SCID mice. Significance among groups was determined by ANOVA. (D) The expression of PD-1 on CD8+ T cells were upregulated in the spleen in decitabine treated ITP mice (D, 2.10 ± 0.65 vs. 3.18 ± 0.92, n = 6, P = 0.039, Unpaired t-test). (E) Increased platelet count was observed after low-dose decitabine treatment in ITP patients (17.08 × 109 ± 8.53 × 109/L vs. 70.75 × 109 ± 47.79 × 109/L, n = 12, P = 0.004, Paired t-test). (F,G) Elevated PD-1 expression on CD8+ T cells was observed after low-dose decitabine treatment in ITP patients (G, 1.24 ± 1.00 vs. 3.62 ± 2.28, n = 12, P = 0.018, Paired t-test). (H) Linear regression analysis of PD-1 expression and platelet counts in active ITP mice on day 28 after decitabine therapy (PBS group and decitabine group). (I) Linear regression analysis of the PD-1 expression on CD8+ T cells and platelet counts in ITP patients before and after decitabine treatment. *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 5
Figure 5
Low-dose decitabine (Dec) decreased PD-1 promoter methylation in ITP in vitro. (A) The representative plots of bisulfite sequencing analysis of PD-1 methylation. (B) Low-dose decitabine significantly decreased the percentage of methylated CpG residues of PD-1 in CD8+ T cells (position 2, 14.00 ± 5.48% vs. 4.00 ± 5.48%, P = 0.020; position 3, 22.00 ± 8.37% vs. 10.00 ± 7.07%, P = 0.040, position 3, 26.00 ± 8.94% vs. 12.00 ± 8.47%, P = 0.034, n = 5, Multiple t-test). *P < 0.05.
Figure 6
Figure 6
Low-dose decitabine (Dec) inhibited CTLs-mediated platelet apoptosis in ITP. (A) The representative dot-plots characterized the apoptosis of platelets treated with PBS or 100 nM decitabine and cultured with CTLs of ITP patients treated with PBS or 100 nM decitabine. (B) A significant reduction in CTLs induced apoptosis of platelets cultured with low-dose decitabine was observed (15.40 ± 8.23 vs. 14.05 ± 7.32, n = 15, P = 0.018, Paired t-test). (C–E) Platelet spontaneous apoptosis and platelet apoptosis cultured with CTLs was measured before (C) and after (D) 12-week decitabine treatment for ITP patients. (E) CTLs-induced platelet apoptosis was significantly inhibited after 12-week decitabine administration (before: median: 4.98, IQR: 2.66–13.95 vs. after: median: 0.61, IQR: 0.37–5.34, n = 6, P = 0.031, Wilcoxon test). (F) Isolated CD8+ T cells were treated with PBS control or decitabine. Western blotting was performed on cell lysates. (G,H) Graphs present densitometry data of relative phosphorylated (p-) protein to total protein after PBS or 100 nM decitabine treatment (G, p-PI3K, 0.82 ± 0.095 vs. 0.53 ± 0.03, P = 0.017; H, p-AKT, 0.75 ± 0.035 vs. 0.50 ± 0.036, n = 3, P = 0.035, Paired t-test). *P < 0.05.
Figure 7
Figure 7
Blocking PD-1 can partly counteract the therapeutic effect of low-dose decitabine in inhibiting CTLs-mediated platelet apoptosis in ITP in vitro and in vivo. (A) Anti-human PD-1 antibody (Anti-hPD-1) partly counteracted decitabine effect in inhibiting CTLs-induced platelets apoptosis in ITP in vitro (13.63 ± 8.05 at Anti-hPD-1 vs. 13.12 ± 6.53 at Dec + Anti-hPD-1, n = 16, P = 0.961; 13.63 ± 8.05 at Anti-hPD-1 vs. 11.86 ± 6.48 at Dec, n = 16, P = 0.043, ANOVA). (B) Anti-mouse PD-1 antibody (Anti-mPD-1) offset the low-dose decitabine treatment in active ITP mice on days 21 and 28 (day 21, 595.0 × 109 ± 55.42 × 109/L at Dec vs. 396.7 × 109 ± 42.40 × 109/L at Dec + Anti-mPD-1, P = 0.026; day 28, 685.0 × 109 ± 51.03 × 109/L at Dec vs. 476.7 × 109 ± 27.16 × 109/L at Dec + Anti-mPD-1, n = 6, P = 0.008, ANOVA). *P < 0.05, **P < 0.01.

References

    1. Cines DB, Bussel JB, Liebman HA, Luning Prak ET. The ITP syndrome: pathogenic and clinical diversity. Blood. (2009) 113:6511–21. 10.1182/blood-2009-01-129155
    1. Cines DB, Cuker A, Semple JW. Pathogenesis of immune thrombocytopenia. Presse Med. (2014) 43:e49–59. 10.1016/j.lpm.2014.01.010
    1. Cooper N. State of the art – how I manage immune thrombocytopenia. Br J Haematol. (2017) 177:39–54. 10.1111/bjh.14515
    1. Miltiadous O, Hou M, Bussel J. Identifying and treating refractory ITP: difficulty in diagnosis and role of combination treatment. Blood. (2020) 135:472–90. 10.1182/blood.2019003599
    1. Olsson B, Andersson PO, Jernas M, Jacobsson S, Carlsson B, Carlsson LM, et al. . T-cell-mediated cytotoxicity toward platelets in chronic idiopathic thrombocytopenic purpura. Nat Med. (2003) 9:1123–4. 10.1038/nm921
    1. Zhang F, Chu X, Wang L, Zhu Y, Li L, Ma D, et al. . Cell-mediated lysis of autologous platelets in chronic idiopathic thrombocytopenic purpura. Eur J Haematol. (2006) 76:427–31. 10.1111/j.1600-0609.2005.00622.x
    1. Zhou H, Qiu JH, Wang T, Yu YY, Liu XN, Li X, et al. . Interleukin 27 inhibits cytotoxic T-lymphocyte-mediated platelet destruction in primary immune thrombocytopenia. Blood. (2014) 124:3316–9. 10.1182/blood-2014-06-580084
    1. Zufferey A, Kapur R, Semple JW. Pathogenesis and therapeutic mechanisms in immune thrombocytopenia (ITP). J Clin Med. (2017) 6:16. 10.3390/jcm6020016
    1. Zhang J, Ma D, Zhu X, Qu X, Ji C, Hou M. Elevated profile of Th17, Th1 and Tc1 cells in patients with immune thrombocytopenic purpura. Haematologica. (2009) 94:1326–9. 10.3324/haematol.2009.007823
    1. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. (1992) 11:3887–95.
    1. Vibhakar R, Juan G, Traganos F, Darzynkiewicz Z, Finger LR. Activation-induced expression of human programmed death-1 gene in T-lymphocytes. Exp Cell Res. (1997) 232:25–8. 10.1006/excr.1997.3493
    1. Dolff S, Quandt D, Feldkamp T, Jun C, Mitchell A, Hua F, et al. . Increased percentages of PD-1 on CD4+ T cells is associated with higher INF-gamma production and altered IL-17 production in patients with systemic lupus erythematosus. Scand J Rheumatol. (2014) 43:307–13. 10.3109/03009742.2013.869830
    1. Guo Y, Walsh AM, Canavan M, Wechalekar MD, Cole S, Yin X, et al. . Immune checkpoint inhibitor PD-1 pathway is down-regulated in synovium at various stages of rheumatoid arthritis disease progression. PLoS ONE. (2018) 13:e0192704. 10.1371/journal.pone.0192704
    1. Cao Q, Zheng C, Xie Z, Liu L, Zhu J, Jin T. The change of PD1, PDL1 in experimental autoimmune encephalomyelitis treated by 1,25(OH)2D3. J Neuroimmunol. (2020) 338:577079. 10.1016/j.jneuroim.2019.577079
    1. Zhong J, Chen S, Xu L, Lai J, Liao Z, Zhang T, et al. . Lower expression of PD-1 and PD-L1 in peripheral blood from patients with chronic ITP. Hematology. (2016) 21:552–7. 10.1080/10245332.2016.1155347
    1. Zhao Y, Han P, Liu L, Wang X, Xu P, Wang H, et al. . Indirubin modulates CD4(+) T-cell homeostasis via PD1/PTEN/AKT signalling pathway in immune thrombocytopenia. J Cell Mol Med. (2019) 23:1885–98. 10.1111/jcmm.14089
    1. Nie M, Liu Y, Li XX, Min YN, Yang DD, Li Q, et al. . PD-1/PD-L Pathway Potentially Involved in ITP Immunopathogenesis. Thromb Haemost. (2019) 119:758–65. 10.1055/s-0039-1679909
    1. Wang J, Yi Z, Wang S, Li Z. The effect of decitabine on megakaryocyte maturation and platelet release. Thromb Haemost. (2011) 106:337–43. 10.1160/TH10-11-0744
    1. Zhou H, Hou Y, Liu X, Qiu J, Feng Q, Wang Y, et al. . Low-dose decitabine promotes megakaryocyte maturation and platelet production in healthy controls and immune thrombocytopenia. Thromb Haemost. (2015) 113:1021–34. 10.1160/TH14-04-0342
    1. Zhou H, Qin P, Liu Q, Yuan C, Hao Y, Zhang H, et al. . A prospective, multicenter study of low dose decitabine in adult patients with refractory immune thrombocytopenia. Am J Hematol. (2019) 94:1374–81. 10.1002/ajh.25646
    1. Orskov AD, Treppendahl MB, Skovbo A, Holm MS, Friis LS, Hokland M, et al. . Hypomethylation and up-regulation of PD-1 in T cells by azacytidine in MDS/AML patients: A rationale for combined targeting of PD-1 and DNA methylation. Oncotarget. (2015) 6:9612–26. 10.18632/oncotarget.3324
    1. Rodeghiero F, Stasi R, Gernsheimer T, Michel M, Provan D, Arnold DM, et al. . Standardization of terminology, definitions and outcome criteria in immune thrombocytopenic purpura of adults and children: report from an international working group. Blood. (2009) 113:2386–93. 10.1182/blood-2008-07-162503
    1. Chow L, Aslam R, Speck E, Kim M, Cridland N, Webster M, et al. . A murine model of severe immune thrombocytopenia is induced by antibody- and CD8+ T cell-mediated responses that are differentially sensitive to therapy. Blood. (2010) 115:1247–53. 10.1182/blood-2009-09-244772
    1. Hou Y, Feng Q, Xu M, Li GS, Liu XN, Sheng Z, et al. . High-dose dexamethasone corrects impaired myeloid-derived suppressor cell function via Ets1 in immune thrombocytopenia. Blood. (2016) 127:1587–97. 10.1182/blood-2015-10-674531
    1. Finlay D, Cantrell DA. Metabolism, migration and memory in cytotoxic T cells. Nat Rev Immunol. (2011) 11:109–17. 10.1038/nri2888
    1. Jin H, Ahmed R, Okazaki T. Role of PD-1 in regulating T-cell immunity. Curr Top Microbiol Immunol. (2011) 350:17–37. 10.1007/82_2010_116
    1. Liu Y, Liu S, Wu C, Huang W, Xu B, Lian S, et al. . In vitro PD-1-Mediated PI3K/Akt/mTOR, Caspase 9/Caspase 3 and ERK pathways are involved in regulating the apoptosis and proliferation of CD4 and CD8 T cells during BVDV infection. Front Immunol. (2020) 11:467. 10.3389/fimmu.2020.00467
    1. Zhao C, Li X, Zhang F, Wang L, Peng J, Hou M. Increased cytotoxic T-lymphocyte-mediated cytotoxicity predominant in patients with idiopathic thrombocytopenic purpura without platelet autoantibodies. Haematologica. (2008) 93:1428–30. 10.3324/haematol.12889
    1. Olsson B, Jernas M, Wadenvik H. Increased plasma levels of granzymes in adult patients with chronic immune thrombocytopenia. Thromb Haemost. (2012) 107:1182–4. 10.1160/TH12-01-0012
    1. Qiu J, Liu X, Li X, Zhang X, Han P, Zhou H, et al. . CD8(+) T cells induce platelet clearance in the liver via platelet desialylation in immune thrombocytopenia. Sci Rep. (2016) 6:27445. 10.1038/srep27445
    1. Chinai J, Janakiram M, Chen F, Chen W, Kaplan M, Zang X. New immunotherapies targeting the PD-1 pathway. Trends Pharmacol Sci. (2015) 36:587–95. 10.1016/j.tips.2015.06.005
    1. Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. (2015) 36:265–76. 10.1016/j.it.2015.02.008
    1. Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, et al. . Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. (2001) 291:319–22. 10.1126/science.291.5502.319
    1. Okazaki T, Tanaka Y, Nishio R, Mitsuiye T, Mizoguchi A, Wang J, et al. . Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med. (2003) 9:1477–83. 10.1038/nm955
    1. Salama AD, Chitnis T, Imitola J, Ansari MJ, Akiba H, Tushima F, et al. . Critical role of the programmed death-1 (PD-1) pathway in regulation of experimental autoimmune encephalomyelitis. J Exp Med. (2003) 198:71–8. 10.1084/jem.20022119
    1. Ansari MJ, Salama AD, Chitnis T, Smith RN, Yagita H, Akiba H, et al. . The programmed death-1 (PD-1) pathway regulates autoimmune diabetes in nonobese diabetic (NOD) mice. J Exp Med. (2003) 198:63–9. 10.1084/jem.20022125
    1. Guleria I, Gubbels Bupp M, Dada S, Fife B, Tang Q, Ansari MJ, et al. . Mechanisms of PDL1-mediated regulation of autoimmune diabetes. Clin Immunol. (2007) 125:16–25. 10.1016/j.clim.2007.05.013
    1. Kanameishi S, Otsuka A, Nonomura Y, Fujisawa A, Endo Y, Kabashima K. Idiopathic thrombocytopenic purpura induced by nivolumab in a metastatic melanoma patient with elevated PD-1 expression on B cells. Ann Oncol. (2016) 27:546–7. 10.1093/annonc/mdv580
    1. Le Roy A, Kempf E, Ackermann F, Routier E, Robert C, Turpin A, et al. . Two cases of immune thrombocytopenia associated with pembrolizumab. Eur J Cancer. (2016) 54:172–4. 10.1016/j.ejca.2015.10.073
    1. Wu D, Liu Y, Pang N, Sun M, Wang X, Haridia Y, et al. . PD-1/PD-L1 pathway activation restores the imbalance of Th1/Th2 and treg/Th17 cells subtypes in immune thrombocytopenic purpura patients. Medicine (Baltimore). (2019) 98:e17608. 10.1097/MD.0000000000017608
    1. Raptopoulou A, Bertsias G, Makrygiannakis D, Verginis P, Kritikos I, Tzardi M, et al. . The programmed death 1/programmed death ligand 1 inhibitory pathway is up-regulated in rheumatoid synovium and regulates peripheral T cell responses in human and murine arthritis. Arthritis Rheum. (2010) 62:1870–80. 10.1002/art.27500
    1. Zhou H, Xiong L, Wang Y, Ding L, Hu S, Zhao M, et al. . Treatment of murine lupus with PD-LIg. Clin Immunol. (2016) 162:1–8. 10.1016/j.clim.2015.10.006
    1. Li H, Xuan M, Yang R. DNA methylation and primary immune thrombocytopenia. Semin Hematol. (2013) 50:S116–26. 10.1053/j.seminhematol.2013.03.022
    1. Zhao H, Xue F, Xu J, Fang Z. Aberrant histone methylation in the patients with immune thrombocytopenia. Platelets. (2014) 25:207–10. 10.3109/09537104.2013.859664
    1. Tao J, Yang M, Chen Z, Huang Y, Zhao Q, Xu J, et al. . Decreased DNA methyltransferase 3A and 3B mRNA expression in peripheral blood mononuclear cells and increased plasma SAH concentration in adult patients with idiopathic thrombocytopenic purpura. J Clin Immunol. (2008) 28:432–9. 10.1007/s10875-008-9223-2
    1. de Vos D, van Overveld W. Decitabine: a historical review of the development of an epigenetic drug. Ann Hematol. (2005) 84:3–8. 10.1007/s00277-005-0008-x
    1. Momparler RL. Pharmacology of 5-Aza-2′-deoxycytidine (decitabine). Semin Hematol. (2005) 42:S9–16. 10.1053/j.seminhematol.2005.05.002
    1. Yang H, Bueso-Ramos C, DiNardo C, Estecio MR, Davanlou M, Geng QR, et al. . Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. (2014) 28:1280–8. 10.1038/leu.2013.355
    1. Chikuma S, Terawaki S, Hayashi T, Nabeshima R, Yoshida T, Shibayama S, et al. . PD-1-mediated suppression of IL-2 production induces CD8+ T cell anergy in vivo. J Immunol. (2009) 182:6682–9. 10.4049/jimmunol.0900080
    1. Zhao R, Song Y, Wang Y, Huang Y, Li Z, Cui Y, et al. . PD-1/PD-L1 blockade rescue exhausted CD8+ T cells in gastrointestinal stromal tumours via the PI3K/Akt/mTOR signalling pathway. Cell Prolif. (2019) 52:e12571. 10.1111/cpr.12571
    1. Jones R, Thompson C. Revving the engine: signal transduction fuels T cell activation. Immunity. (2007) 27:173–8. 10.1016/j.immuni.2007.07.008
    1. Wang R, Dillon C, Shi L, Milasta S, Carter R, Finkelstein D, et al. . The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity. (2011) 35:871–82. 10.1016/j.immuni.2011.09.021

Source: PubMed

3
Abonneren