Low-dose decitabine modulates T-cell homeostasis and restores immune tolerance in immune thrombocytopenia

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

Abstract

Our previous clinical study showed that low-dose decitabine exhibited sustained responses in nearly half of patients with refractory immune thrombocytopenia (ITP). The long-term efficacy of decitabine in ITP is not likely due to its simple role in increasing platelet production. Whether decitabine has the potential to restore immune tolerance in ITP is unknown. In this study, we analyzed the effect of decitabine on T-cell subpopulations in ITP in vitro and in vivo. We found that low-dose decitabine promoted the generation and differentiation of regulatory T (Treg) cells and augmented their immunosuppressive function. Splenocytes from CD61 knockout mice immunized with CD61+ platelets were transferred into severe combined immunodeficient mouse recipients to induce a murine model of ITP. Low-dose decitabine alleviated thrombocytopenia and restored the balance between Treg and helper T (Th) cells in active ITP mice. Treg deletion and depletion offset the effect of decitabine in restoring CD4+ T-cell subpopulations in ITP mice. For patients who received low-dose decitabine, the quantity and function of Treg cells were substantially improved, whereas Th1 and Th17 cells were suppressed compared with the pretreatment levels. Next-generation RNA-sequencing and cytokine analysis showed that low-dose decitabine rebalanced T-cell homeostasis, decreased proinflammatory cytokines, and downregulated phosphorylated STAT3 in patients with ITP. STAT3 inhibition analysis suggested that low-dose decitabine might restore Treg cells by inhibiting STAT3 activation. In conclusion, our data indicate that the immunomodulatory effect of decitabine provides one possible mechanistic explanation for the sustained response achieved by low-dose decitabine in ITP.

© 2021 by The American Society of Hematology.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Low-dose decitabine had no influence on the apoptosis of PBMCs from patients with ITP or healthy control subjects, but significantly elevated the percentage of Treg cells in patients with ITP without changing the percentage of CD4+ T cells in PBMCs. (A) Representative dot plots of flow cytometry analysis of apoptosis in PBMCs from patients with ITP. The percentage of Annexin-positive and propidium iodide (PI)-negative cells in PBMCs showed the cell apoptosis rate. (B,C) Decitabine induced apoptosis of PBMCs from healthy control subjects and patients with ITP at doses of 1 μM and 10 μM (n = 6). (D-F) Percentages of Treg cells in CD4+ T cells were significantly increased by decitabine at the doses of 10 nM and 100 nM in patients with ITP (n = 11). (G-I) In cultured isolated CD4+ T cells, low-dose decitabine (100 nM) significantly increased the percentage of Treg cells in patients with ITP (I; n = 7), but not in healthy control subjects (H; n = 8). *P < .05, **P < .01. Dec, decitabine; HC, healthy controls; ns, not significant.
Figure 2.
Figure 2.
The inhibitory function of Treg cells was enhanced by low-dose decitabine in ITP in vitro. Teff cells (2 × 105 cells per well) were seeded in a 96-well plate with or without Treg cells (5 × 104 cells per well) in the presence of decitabine 100 nM. The cell division index was calculated based on the dilution of CFSE fluorescence measured by flow cytometry and represents the average number of cell divisions that Teff cells in the original population have undergone [division index = sum (i × N(i)/2i)/sum (N(i)/2i), “i” is division number (undivided = 0), and “N(i)” is the number of cells in division “i”]. (A,C) Representative histograms of CD4+CFSE+ Teff cells from one healthy control subject (A) and patient with ITP (C). (B,D) Treg cells suppressed the proliferation of Teff cells. Decitabine (100 nM) enhanced the inhibitory function of Treg cells in ITP (D; n = 24), but not in healthy control subjects (B; n = 13). *P < .05, **P < .01, ***P < .001. Dec, decitabine; HC, healthy control subjects; ns, not significant.
Figure 3.
Figure 3.
Low-dose decitabine ameliorated thrombocytopenia in active ITP murine models via Treg cells. ITP models were established in irradiated SCID mice with engraftment of 2 × 104 splenocytes from CD61 knockout mice immunized against wild-type C57 mice platelets; platelet counts were then monitored every week for 4 weeks (mean ± standard error of the mean). (A) From day 7, decitabine (0, 0.01 mg/kg, 0.03 mg/kg) was administered. The horizontally dotted lines represent the baseline platelet counts of SCID mice (mean ± standard error of the mean). On days 21 and 28, significantly higher platelet counts were observed in the group administered the 0.03-mg/kg dose of decitabine compared with control group. (B) Decitabine-treated ITP mice had a significantly higher percentage of splenic Treg cells in CD4+ T cells compared with control mice (n = 11 in the control group and n = 13 in the decitabine group, respectively). The percentage of Th1 (C) and Th17 (D) in CD4+ T cells were decreased in decitabine-treated spleens compared with controls. No significance was found in Th22 cells (E) in splenic CD4+ T cells (n = 9 in the control group and n = 10 in the decitabine group). Treg deletion (F) by magnetic beads and depletion (K) by anti-CD25 antibody partly offset the effect of decitabine on increasing platelet counts of ITP mice (n = 6). Treg-deleted splenocyte-transferred mice (G) and anti-CD25 antibody-treated mice (L) were significantly depleted of Treg cells in the spleens on day 28 (n = 6). (H-J) After Treg deletion, decitabine had no significant effect on splenic Th1, Th17, or Th22 cells on day 28 in ITP mice (n = 6). (M-O) With Treg depletion by anti-CD25 antibody, decitabine had no significant effect on splenic Th1, Th17, or Th22 cells on day 28 in ITP mice (n = 6). *P < .05, **P < .01, ***P < .001. Dec, decitabine; ns, not significant.
Figure 4.
Figure 4.
Immunologic responses to low-dose decitabine in patients with ITP. (A-E) Percentages of Treg, Th1, Th17, Th2, and Th22 cells in peripheral blood were analyzed by using flow cytometry. Treg cells (n = 22), Th1 cells (n = 17), Th17 cells (n = 15), Th2 cells (n = 8), and Th22 cells (n = 16) in CD4+ T cells before and after decitabine treatment are shown. (F) Ex vivo suppression assays were performed to examine the suppressive activity of Treg cells obtained from patients with ITP before and after decitabine treatment. CFSE-labeled CD4+CD25– Teff cells were stimulated and cocultured with Treg cells from each patient. Representative histograms show proliferation of CD4+ Teff cells with or without Treg cells before (above) or after (below) decitabine treatment. (G-H) Treg cell–mediated suppression of CD4+ Teff cell proliferation was measured using division index before and after decitabine treatment (n = 14). (I) Decitabine enhanced immunosuppressive function of Treg cells. *P < .05, **P < .01, ****P < .0001. NR, nonresponders; R, responders.
Figure 5.
Figure 5.
Profiles and enriched pathway analysis of decitabine-induced genes. (A) Venn diagram of overlapping genes of decitabine-altered genes in 4 responders. (B) Heat map of decitabine-altered genes in 4 responders with unsupervised clustering. Red to green colors represent relatively high to low gene expression, respectively, and gray indicates below the detectable level. (C) Venn diagram of overlapping genes of decitabine-altered genes in 3 nonresponders. (D) No overlap of decitabine-altered genes was observed between responders and nonresponders. (E-G) Pathways were predicted by Gene Ontology (E), KEGG (F), and Reactome (G) databases based on the expression changes of decitabine altered genes in 4 responders. AF, after; BE, before; NR, nonresponders; R, responders.
Figure 6.
Figure 6.
Low-dose decitabine inhibited the STAT3 signaling pathway. (A) Representative western blots of phosphorylated-STAT3 (p-STAT3), total STAT, phosphorylated-AKT (p-AKT), total AKT, and GAPDH in PBMCs from patients with ITP before and after decitabine treatment. (B-C) Graphs present densitometry data of relative phosphorylated protein to total protein (n = 3). Treg cells in CD4+ T cells from healthy control subjects (D; n = 10) and patients with ITP (E; n = 8) in the STAT3 inhibitor plus decitabine group were no different from those in the STAT3 inhibitor group in vitro. In the presence of AKT inhibitor, decitabine significantly increased the percentage of Treg cells from patients with ITP in vitro (G; n = 10), but not in the healthy control subjects (F; n = 8). (H) The platelet counts in the STAT3 inhibitor plus decitabine group were not significantly different from those in mice receiving STAT3 inhibitor (n = 6). (I) AKT inhibitor plus decitabine increased platelet counts in mice compared with AKT inhibitor on day 21 (n = 6). *P < .05. Dec, decitabine; ns, not significant.

References

    1. Olsson B, Andersson PO, Jernås M, et al. . T-cell-mediated cytotoxicity toward platelets in chronic idiopathic thrombocytopenic purpura. Nat Med. 2003;9(9):1123-1124.
    1. McMillan R, Wang L, Tomer A, Nichol J, Pistillo J.. Suppression of in vitro megakaryocyte production by antiplatelet autoantibodies from adult patients with chronic ITP. Blood. 2004;103(4):1364-1369.
    1. Cines DB, Bussel JB, Liebman HA, Luning Prak ET.. The ITP syndrome: pathogenic and clinical diversity. Blood. 2009;113(26):6511-6521.
    1. Kuter DJ, Rummel M, Boccia R, et al. . Romiplostim or standard of care in patients with immune thrombocytopenia. N Engl J Med. 2010;363(20):1889-1899.
    1. Imbach P, Crowther M.. Thrombopoietin-receptor agonists for primary immune thrombocytopenia. N Engl J Med. 2011;365(8):734-741.
    1. Cines DB, Cuker A, Semple JW.. Pathogenesis of immune thrombocytopenia. Presse Med. 2014;43(4 pt 2):e49-e59.
    1. Nishimoto T, Kuwana M.. CD4+CD25+Foxp3+ regulatory T cells in the pathophysiology of immune thrombocytopenia. Semin Hematol. 2013; 50(suppl 1):S43-S49.
    1. Son BR, Kim JY.. Association of CD4(+)CD25(+)FoxP3(+) regulatory T cells with natural course of childhood chronic immune thrombocytopenic purpura. Korean J Pediatr. 2015;58(5):178-182.
    1. Arandi N, Mirshafiey A, Jeddi-Tehrani M, et al. . Alteration in frequency and function of CD4+CD25+FOXP3+ regulatory T cells in patients with immune thrombocytopenic purpura. Iran J Allergy Asthma Immunol. 2014;13(2):85-92.
    1. Panitsas FP, Theodoropoulou M, Kouraklis A, et al. . Adult chronic idiopathic thrombocytopenic purpura (ITP) is the manifestation of a type-1 polarized immune response. Blood. 2004;103(7):2645-2647.
    1. McKenzie CGJ, Guo L, Freedman J, Semple JW.. Cellular immune dysfunction in immune thrombocytopenia (ITP). Br J Haematol. 2013;163(1):10-23.
    1. Li J, Wang Z, Hu S, Zhao X, Cao L.. Correction of abnormal T cell subsets by high-dose dexamethasone in patients with chronic idiopathic thrombocytopenic purpura. Immunol Lett. 2013;154(1-2):42-48.
    1. Li Z, Mou W, Lu G, et al. . Low-dose rituximab combined with short-term glucocorticoids up-regulates Treg cell levels in patients with immune thrombocytopenia. Int J Hematol. 2011;93(1):91-98.
    1. Bao W, Bussel JB, Heck S, et al. . Improved regulatory T-cell activity in patients with chronic immune thrombocytopenia treated with thrombopoietic agents. Blood. 2010;116(22):4639-4645.
    1. Steensma DP.Myelodysplasia, megakaryocytes, and methylation. Leuk Res. 2004;28(8):775-776.
    1. Wang J, Yi Z, Wang S, Li Z.. The effect of decitabine on megakaryocyte maturation and platelet release. Thromb Haemost. 2011;106(2):337-343.
    1. Zhou H, Hou Y, Liu X, et al. . Low-dose decitabine promotes megakaryocyte maturation and platelet production in healthy controls and immune thrombocytopenia. Thromb Haemost. 2015;113(5):1021-1034.
    1. Zhou H, Qin P, Liu Q, et al. . A prospective, multicenter study of low dose decitabine in adult patients with refractory immune thrombocytopenia. Am J Hematol. 2019;94(12):1374-1381.
    1. Kim HP, Leonard WJ.. CREB/ATF-dependent T cell receptor-induced FoxP3 gene expression: a role for DNA methylation. J Exp Med. 2007;204(7):1543-1551.
    1. Sánchez-Abarca LI, Gutierrez-Cosio S, Santamaría C, et al. . Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting. Blood. 2010;115(1):107-121.
    1. Chow L, Aslam R, Speck ER, 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(6):1247-1253.
    1. Hou Y, Feng Q, Xu M, et al. . High-dose dexamethasone corrects impaired myeloid-derived suppressor cell function via Ets1 in immune thrombocytopenia. Blood. 2016;127(12):1587-1597.
    1. Aslam R, Hu Y, Gebremeskel S, et al. . Thymic retention of CD4+CD25+FoxP3+ T regulatory cells is associated with their peripheral deficiency and thrombocytopenia in a murine model of immune thrombocytopenia. Blood. 2012;120(10):2127-2132.
    1. Liu W, Li H, Hao Y, et al. . Decreased immunosuppressive actions of 1α, 25-dihydroxyvitamin D3 in patients with immune thrombocytopenia. Mol Immunol. 2016;78:89-97.
    1. Chen M, Yan R, Zhou K, et al. . Akt-mediated platelet apoptosis and its therapeutic implications in immune thrombocytopenia. Proc Natl Acad Sci U S A. 2018;115(45):E10682-E10691.
    1. Hu Y, Wang X, Yu S, Hou Y, Ma D, Hou M.. Neutralizations of IL-17A and IL-21 regulate regulatory T cell/T-helper 17 imbalance via T-helper 17-associated signaling pathway in immune thrombocytopenia. Expert Opin Ther Targets. 2015;19(6):723-732.
    1. Cuker A, Neunert CE.. How I treat refractory immune thrombocytopenia. Blood. 2016;128(12):1547-1554.
    1. Lambert MP, Gernsheimer TB.. Clinical updates in adult immune thrombocytopenia. Blood. 2017;129(21):2829-2835.
    1. Li H, Xuan M, Yang R.. DNA methylation and primary immune thrombocytopenia. Semin Hematol. 2013;50(suppl 1):S116-S126.
    1. Chen Z, Guo Z, Ma J, Ma J, Liu F, Wu R.. Foxp3 methylation status in children with primary immune thrombocytopenia. Hum Immunol. 2014;75(11):1115-1119.
    1. Chen ZP, Gu DS, Zhou ZP, et al. . Decreased expression of MBD2 and MBD4 gene and genomic-wide hypomethylation in patients with primary immune thrombocytopenia. Hum Immunol. 2011;72(6):486-491.
    1. Han P, Yu T, Hou Y, et al. . Low-dose decitabine inhibits cytotoxic T lymphocytes-mediated platelet destruction via modulating PD-1 methylation in immune thrombocytopenia. Front Immunol. 2021;12:630693.
    1. Wang L, Amoozgar Z, Huang J, et al. . Decitabine enhances lymphocyte migration and function and synergizes with CTLA-4 blockade in a murine ovarian cancer model. Cancer Immunol Res. 2015;3(9):1030-1041.
    1. Choi J, Ritchey J, Prior JL, et al. . In vivo administration of hypomethylating agents mitigate graft-versus-host disease without sacrificing graft-versus-leukemia. Blood. 2010;116(1):129-139.
    1. Mangano K, Fagone P, Bendtzen K, et al. . Hypomethylating agent 5-aza-2'-deoxycytidine (DAC) ameliorates multiple sclerosis in mouse models. J Cell Physiol. 2014;229(12):1918-1925.
    1. Wang X, Wang J, Yu Y, et al. . Decitabine inhibits T cell proliferation via a novel TET2-dependent mechanism and exerts potent protective effect in mouse auto- and allo-immunity models. Oncotarget. 2017;8(34):56802-56815.
    1. Zheng Q, Xu Y, Liu Y, et al. . Induction of Foxp3 demethylation increases regulatory CD4+CD25+ T cells and prevents the occurrence of diabetes in mice. J Mol Med (Berl). 2009;87(12):1191-1205.
    1. Fagone P, Mazzon E, Chikovani T, et al. . Decitabine induces regulatory T cells, inhibits the production of IFN-gamma and IL-17 and exerts preventive and therapeutic efficacy in rodent experimental autoimmune neuritis. J Neuroimmunol. 2018;321:41-48.
    1. Guo L, Kapur R, Aslam R, et al. . CD20+ B-cell depletion therapy suppresses murine CD8+ T-cell-mediated immune thrombocytopenia. Blood. 2016;127(6):735-738.
    1. Stasi R, Cooper N, Del Poeta G, et al. . Analysis of regulatory T-cell changes in patients with idiopathic thrombocytopenic purpura receiving B cell-depleting therapy with rituximab. Blood. 2008;112(4):1147-1150.
    1. Ling Y, Cao X, Yu Z, Ruan C.. Circulating dendritic cells subsets and CD4+Foxp3+ regulatory T cells in adult patients with chronic ITP before and after treatment with high-dose dexamethasome. Eur J Haematol. 2007;79(4):310-316.
    1. Wang SC, Yang KD, Lin CY, et al. . Intravenous immunoglobulin therapy enhances suppressive regulatory T cells and decreases innate lymphoid cells in children with immune thrombocytopenia. Pediatr Blood Cancer. 2020;67(2):e28075.
    1. Guo L, Yang L, Speck ER, et al. . Allogeneic platelet transfusions prevent murine T-cell-mediated immune thrombocytopenia. Blood. 2014;123(3):422-427.
    1. Semple JW, Milev Y, Cosgrave D, et al. . Differences in serum cytokine levels in acute and chronic autoimmune thrombocytopenic purpura: relationship to platelet phenotype and antiplatelet T-cell reactivity. Blood. 1996;87(10):4245-4254.
    1. Qu MM, Liu XN, Liu XG, et al. . Cytokine changes in response to TPO receptor agonist treatment in primary immune thrombocytopenia. Cytokine. 2017;92:110-117.
    1. Andersson PO, Olsson A, Wadenvik H.. Reduced transforming growth factor-beta1 production by mononuclear cells from patients with active chronic idiopathic thrombocytopenic purpura. Br J Haematol. 2002;116(4):862-867.
    1. Guo C, Chu X, Shi Y, et al. . Correction of Th1-dominant cytokine profiles by high-dose dexamethasone in patients with chronic idiopathic thrombocytopenic purpura. J Clin Immunol. 2007;27(6):557-562.
    1. Villarino AV, Kanno Y, O’Shea JJ.. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat Immunol. 2017;18(4):374-384.
    1. Laurence A, Amarnath S, Mariotti J, et al. . STAT3 transcription factor promotes instability of nTreg cells and limits generation of iTreg cells during acute murine graft-versus-host disease. Immunity. 2012;37(2):209-222.
    1. Burchill MA, Yang J, Vogtenhuber C, Blazar BR, Farrar MA.. IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells. J Immunol. 2007;178(1):280-290.
    1. Yao Z, Kanno Y, Kerenyi M, et al. . Nonredundant roles for Stat5a/b in directly regulating Foxp3. Blood. 2007;109(10):4368-4375.
    1. Zorn E, Nelson EA, Mohseni M, et al. . IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood. 2006;108(5):1571-1579.
    1. Fujino M, Li XK.. Role of STAT3 in regulatory T lymphocyte plasticity during acute graft-vs.-host-disease. JAK-STAT. 2013;2(4):e24529.
    1. Sailer T, Lechner K, Panzer S, Kyrle PA, Pabinger I.. The course of severe autoimmune thrombocytopenia in patients not undergoing splenectomy. Haematologica. 2006;91(8):1041-1045.
    1. Schifferli A, Holbro A, Chitlur M, et al. ; Intercontinental Cooperative ITP Study Group (ICIS). A comparative prospective observational study of children and adults with immune thrombocytopenia: 2-year follow-up. Am J Hematol. 2018;93(6):751-759.
    1. Neunert C, Terrell DR, Arnold DM, et al. . American Society of Hematology 2019 guidelines for immune thrombocytopenia [published correction appears in Blood Adv. 2020;4(2):252]. Blood Adv. 2019;3(23):3829-3866.
    1. Provan D, Arnold DM, Bussel JB, et al. . Updated international consensus report on the investigation and management of primary immune thrombocytopenia. Blood Adv. 2019;3(22):3780-3817.

Source: PubMed

3
Subscribe