Plasmodium infection inhibits the expansion and activation of MDSCs and Tregs in the tumor microenvironment in a murine Lewis lung cancer model

Dickson Adah, Yijun Yang, Quan Liu, Kranthi Gadidasu, Zhu Tao, Songlin Yu, Linglin Dai, Xiaofen Li, Siting Zhao, Limei Qin, Li Qin, Xiaoping Chen, Dickson Adah, Yijun Yang, Quan Liu, Kranthi Gadidasu, Zhu Tao, Songlin Yu, Linglin Dai, Xiaofen Li, Siting Zhao, Limei Qin, Li Qin, Xiaoping Chen

Abstract

Background: A major challenge in the development of effective cancer immunotherapy is the ability of tumors and their microenvironment to suppress immune cells through immunosuppressive cells such as myeloid -derived suppressor cells and regulatory T cells. We previously demonstrated that Plasmodium infection promotes innate and adaptive immunity against cancer in a murine Lewis lung cancer model but its effects on immunosuppressive cells in the tumor microenvironment are unknown.

Methods: Whole Tumors and tumor-derived sorted cells from tumor-bearing mice treated with or without plasmodium infected red blood cells were harvested 17 days post tumor implantation and analyzed using QPCR, western blotting, flow cytometry, and functional assays. Differences between groups were analyzed for statistical significance using Student's t-test.

Results: Here we found that Plasmodium infection significantly reduced the proportions of MDSCs and Tregs in the lung tumor tissues of the treated mice by downregulating their recruiting molecules and blocking cellular activation pathways. Importantly, CD8+ T cells isolated from the tumors of Plasmodium-treated mice exhibited significantly higher levels of granzyme B and perforin and remarkably lower levels of PD-1.

Conclusion: We reveal for the first time, the effects of Plasmodium infection on the expansion and activation of MDSCs and Tregs with a consequent elevation of CD8+T cell-mediated cytotoxicity within the tumor microenvironment and hold great promise for the development of effective immunotherapeutic strategies.

Keywords: Lung cancer; MDSC; PD-1; Recruiting molecules; Tregs.

Conflict of interest statement

Ethics approval

The animal experiment facilities were approved by the Guangdong Provincial Department of Science and Technology, and complied with the guidelines of the Animal Care Committee, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences. All efforts were made to minimize animal suffering.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Downregulation of the population of MDSCs in the tumor microenvironment by Plasmodium. a Gating protocol for the identification of MDSC subsets. b Gated polymorphonuclear and monocytic MDSCs in the tumor tissues of uninfected tumor-bearing mice (LR, control). c Gated polymorphonuclear and monocytic MDSCs in the tumor tissues of Py-infected tumor-bearing mice (treated, LP). d Comparison of the total MDSC population in the tumor tissues of both the treated and the control groups. e The relative proportions of polymorphonuclear and monocytic MDSC subsets in the tumor tissues of tumor-bearing mice treated with infected and uninfected RBCs. f Tumors harvested from tumor-bearing mice treated or untreated with Plasmodium 18 days after tumor inoculation. A total of 50,000 live events was recorded and analyzed per sample. The average number of MDSCs gated out were 2000 cells and 5000 cells for (LP) and (LR) respectively
Fig. 2
Fig. 2
Effects of Plasmodium on the population of Tregs in the tumor microenvironment. a Gating protocol for CD4+ cells. b Gated CD25+FOXP3+ Treg population in untreated tumor tissues. c Gated CD25+FOXP3+ Treg population in treated tumor tissues. d The populations of Tregs in the tumors were compared. Plasmodium infection significantly downregulated the populations of Tregs in the tumor tissues of tumor-bearing mice compared to the control mice. ***P < 0.001
Fig. 3
Fig. 3
Expression levels of key MDSC- and Treg-recruiting cytokines and chemokines in the tumor tissues assessed by qRT-PCR and western blotting. The relative mRNA expression levels of GM-CSF (a), G-CSF (b), M-CSF (c), IL-1β (d), VEGF (e), IL-6 (f), IFN-g (g) IL-14 (h), IL-13 (i), CCL-17 (i) TGF-β (j), CCL-17 (k), and CCL-22 (l) in the tumor tissues of Py-infected tumor-bearing mice compared with those of the control mice. Western blot results of CCR4 protein expression in the tumor tissues (m). β-Actin was used as a loading control. Plasmodium infection significantly decreased the relative mRNA levels of key MDSC and Treg cytokines and chemokines in the tumor tissues of Py-infected tumor-bearing mice compared to the control mice
Fig. 4
Fig. 4
Effects of Plasmodium infection on MDSC signal transduction and downstream protein expression. The expression levels of pSTAT1, pSTAT3, pSTAT5, pSTAT6, NF-κB, Survivin, and S100A9 were assessed by qRT-PCR and western blot analysis. a The protein expression levels of pSTAT1, pSTAT3, pSTAT5, pSTAT6, NF-KB, Survivin and S100A9 in MDSCs from the infected and control mice, as determined by western blotting. b, c qRT-PCR analysis of Survivin and S100A9 expression of the sorted MDSCs. d The protein expression of pSTAT3 in the two MDSC subsets was assessed. (e) Purity of the sorted MDSCs. β-Actin was used as a loading control for both the qRT-PCR and western blot analysis. *P < 0.05. MM, monocytic MDSC; PM, polymorphonuclear MDSC
Fig. 5
Fig. 5
Effects of Plasmodium infection on the expression of immunosuppressive molecules by sorted MDSCs. The MDSC expression of IL-10, arginase 1, NOS2, and ROS was assessed by qRT-PCR and functional assays. Relative mRNA expression levels of IL-10 (a), NOS2 (b), and arginase 1 (c) of MDSCs isolated from the tumor tissues of Py-treated and untreated tumor-bearing mice. The levels of ROS (d) and arginase activity (e) were detected using a DCFDA Cellular ROS Detection Assay Kit (Abcam; cat. # ab113851) and Arginase Activity Assay Kit (Abcam; cat. # ab180877), respectively. **P < 0.01, ****P < 0.0001
Fig. 6
Fig. 6
Effects of Plasmodium infection on CD8+ T cells in the tumor microenvironment. a Flow cytometry gating protocol for CD8+ T cell sorting. b Comparison of the mRNA expression of PD-1 on sorted CD8+ T cells. c mRNA expression of granzyme B (d) and perforin by the sorted CD8+ T cells. e Protein expression of PD-1 on sorted CD8+ T cells determined by western blotting. **P < 0.01
Fig. 7
Fig. 7
Plasmodium infection inhibits tumor cytokines and chemokines secretions through exosomes-like vesicles. To determine whether or not cell-to-cell contact was required for the inhibition of tumor cytokines, we co-cultured LLC cells with either infected RBCs (Li), uninfected RBCs (LRB), exosomes isolated from uninfected mice (LN), exosomes isolated from Py-infected mice (LP), exosomes isolated from tumor-bearing mice infected with Py (LLP), or plasma from Py-infected mice after exosomes have been isolated (LPL). Cells were cultured for either 24 or 48 h and subsequently harvested for RNA extraction and QPCR analysis. Our QPCR results suggest that infected RBCs does not communicate with tumor cells through cell-to-cell contact. Exosome-like vesicles significantly inhibited tumor release of GMCSF(a), IL-10(b), IL-6(c), CCL-17(d), and CCL-22(e) at the mRNA levels

References

    1. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–867. doi: 10.1038/nature01322.
    1. Gattinoni L, Powell DJ, Jr, Rosenberg SA, Restifo NP. Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol. 2006;6:383–393. doi: 10.1038/nri1842.
    1. Li Q, Pan PY, Gu P, Xu D, Chen SH. Role of immature myeloid gr-1+ cells in the development of antitumor immunity. Cancer Res. 2004;64:1130–1139. doi: 10.1158/0008-5472.CAN-03-1715.
    1. Rodriguez PC, et al. Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma. J Exp Med. 2005;202:931–939. doi: 10.1084/jem.20050715.
    1. Serafini P, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med. 2006;203:2691–2702. doi: 10.1084/jem.20061104.
    1. Curiel TJ. Tregs and rethinking cancer immunotherapy. J Clin Invest. 2007;117:1167–1174. doi: 10.1172/JCI31202.
    1. Kusmartsev SA, Li Y, Chen SH. Gr-1+ myeloid cells derived from tumor-bearing mice inhibit primary T cell activation induced through CD3/CD28 costimulation. J Immunol. 2000;165:779–785. doi: 10.4049/jimmunol.165.2.779.
    1. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162–174. doi: 10.1038/nri2506.
    1. Talmadge JE, Gabrilovich DI. History of myeloid-derived suppressor cells. Nat Rev Cancer. 2013;13:739–752. doi: 10.1038/nrc3581.
    1. Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest. 2015;125:3356–3364. doi: 10.1172/JCI80005.
    1. Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest. 2015;125:3365–3376. doi: 10.1172/JCI80006.
    1. Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016;37:208–220. doi: 10.1016/j.it.2016.01.004.
    1. Djeu J, Wei S. Chemoimmunomodulation of MDSCs as a novel strategy for cancer therapy. Oncoimmunology. 2012;1:121–122. doi: 10.4161/onci.1.1.18074.
    1. Pan PY, et al. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010;70:99–108. doi: 10.1158/0008-5472.CAN-09-1882.
    1. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133:775–787. doi: 10.1016/j.cell.2008.05.009.
    1. Nishikawa H, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Curr Opin Immunol. 2014;27:1–7. doi: 10.1016/j.coi.2013.12.005.
    1. Elkord E, et al. T regulatory cells in cancer: recent advances and therapeutic potential. Expert Opin Biol Ther. 2010;10:1573–1586. doi: 10.1517/14712598.2010.529126.
    1. Kakita N, et al. Comparative analyses of regulatory T cell subsets in patients with hepatocellular carcinoma: a crucial role of CD25(−) FOXP3(−) T cells. Int J Cancer. 2012;131:2573–2583. doi: 10.1002/ijc.27535.
    1. Amedei A, et al. Ex vivo analysis of pancreatic cancer-infiltrating T lymphocytes reveals that ENO-specific Tregs accumulate in tumor tissue and inhibit Th1/Th17 effector cell functions. Cancer Immunol Immunother. 2013;62:1249–1260. doi: 10.1007/s00262-013-1429-3.
    1. Yi Y, et al. The functional impairment of HCC-infiltrating gammadelta T cells, partially mediated by regulatory T cells in a TGFbeta- and IL-10-dependent manner. J Hepatol. 2013;58:977–983. doi: 10.1016/j.jhep.2012.12.015.
    1. Scurr M, et al. Highly prevalent colorectal cancer-infiltrating LAP(+) Foxp3(−) T cells exhibit more potent immunosuppressive activity than Foxp3(+) regulatory T cells. Mucosal Immunol. 2014;7:428–439. doi: 10.1038/mi.2013.62.
    1. Chen L, et al. Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity. PLoS One. 2011;6:e24407. doi: 10.1371/journal.pone.0024407.
    1. Roetynck S, et al. Natural killer cells and malaria. Immunol Rev. 2006;214:251–263. doi: 10.1111/j.1600-065X.2006.00446.x.
    1. Kalinski P, et al. Natural killer-dendritic cell cross-talk in cancer immunotherapy. Expert Opin Biol Ther. 2005;5:1303–1315. doi: 10.1517/14712598.5.10.1303.
    1. Woan K, Reddy V. Potential therapeutic role of natural killer cells in cancer. Expert Opin Biol Ther. 2007;7:17–29. doi: 10.1517/14712598.7.1.17.
    1. Ing R, Segura M, Thawani N, Tam M, Stevenson MM. Interaction of mouse dendritic cells and malaria-infected erythrocytes: uptake, maturation, and antigen presentation. J Immunol. 2006;176:441–450. doi: 10.4049/jimmunol.176.1.441.
    1. Corzo CA, et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207:2439–2453. doi: 10.1084/jem.20100587.
    1. Gros A, et al. Myeloid cells obtained from the blood but not from the tumor can suppress T-cell proliferation in patients with melanoma. Clin Cancer Res. 2012;18:5212–5223. doi: 10.1158/1078-0432.CCR-12-1108.
    1. Liu Y, et al. Cell surface receptor FPR2 promotes antitumor host defense by limiting M2 polarization of macrophages. Cancer Res. 2013;73:550–560. doi: 10.1158/0008-5472.CAN-12-2290.
    1. Ondondo B, Jones E, Godkin A, Gallimore A. Home sweet home: the tumor microenvironment as a haven for regulatory T cells. Front Immunol. 2013;4:197. doi: 10.3389/fimmu.2013.00197.
    1. Mailloux AW, Young MR. Regulatory T-cell trafficking: from thymic development to tumor-induced immune suppression. Crit Rev Immunol. 2010;30:435–447. doi: 10.1615/CritRevImmunol.v30.i5.30.
    1. Kortylewski M, et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat Med. 2005;11:1314–1321. doi: 10.1038/nm1325.
    1. Kujawski M, et al. Stat3 mediates myeloid cell-dependent tumor angiogenesis in mice. J Clin Invest. 2008;118:3367–3377. doi: 10.1172/JCI35213.
    1. Amin HM, et al. Selective inhibition of STAT3 induces apoptosis and G(1) cell cycle arrest in ALK-positive anaplastic large cell lymphoma. Oncogene. 2004;23:5426–5434. doi: 10.1038/sj.onc.1207703.
    1. Tsareva SA, et al. Signal transducer and activator of transcription 3 activation promotes invasive growth of colon carcinomas through matrix metalloproteinase induction. Neoplasia. 2007;9:279–291. doi: 10.1593/neo.06820.
    1. Abad C, et al. Targeted STAT3 disruption in myeloid cells alters immunosuppressor cell abundance in a murine model of spontaneous medulloblastoma. J Leukoc Biol. 2014;95:357–367. doi: 10.1189/jlb.1012531.
    1. Ochoa AC, Zea AH, Hernandez C, Rodriguez PC. Arginase, prostaglandins, and myeloid-derived suppressor cells in renal cell carcinoma. Clin Cancer Res. 2007;13:721s–726s. doi: 10.1158/1078-0432.CCR-06-2197.
    1. Riley JL. PD-1 signaling in primary T cells. Immunol Rev. 2009;229:114–125. doi: 10.1111/j.1600-065X.2009.00767.x.
    1. Coppi A, et al. Heparan sulfate proteoglycans provide a signal to Plasmodium sporozoites to stop migrating and productively invade host cells. Cell Host Microbe. 2007;2:316–327. doi: 10.1016/j.chom.2007.10.002.
    1. Bindea G, Mlecnik B, Fridman WH, Pages F, Galon J. Natural immunity to cancer in humans. Curr Opin Immunol. 2010;22:215–222. doi: 10.1016/j.coi.2010.02.006.
    1. Boon T, et al. Identification of tumour rejection antigens recognized by T lymphocytes. Cancer Surv. 1992;13:23–37.
    1. DuPage M, et al. Endogenous T cell responses to antigens expressed in lung adenocarcinomas delay malignant tumor progression. Cancer Cell. 2011;19:72–85. doi: 10.1016/j.ccr.2010.11.011.
    1. Galon J, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–1964. doi: 10.1126/science.1129139.
    1. Parmiani G, De Filippo A, Novellino L, Castelli C. Unique human tumor antigens: immunobiology and use in clinical trials. J Immunol. 2007;178:1975–1979. doi: 10.4049/jimmunol.178.4.1975.
    1. Bronte V, Zanovello P. Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol. 2005;5:641–654. doi: 10.1038/nri1668.
    1. Nagaraj S, Gabrilovich DI. Tumor escape mechanism governed by myeloid-derived suppressor cells. Cancer Res. 2008;68:2561–2563. doi: 10.1158/0008-5472.CAN-07-6229.
    1. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–444. doi: 10.1038/nature07205.
    1. Young MR, Wright MA, Pandit R. Myeloid differentiation treatment to diminish the presence of immune-suppressive CD34+ cells within human head and neck squamous cell carcinomas. J Immunol. 1997;159:990–996.
    1. Kusmartsev SA, Kusmartseva IN, Afanasyev SG, Cherdyntseva NV. Immunosuppressive cells in bone marrow of patients with stomach cancer. Adv Exp Med Biol. 1998;451:189–194. doi: 10.1007/978-1-4615-5357-1_30.
    1. Kusmartsev SA, Ogreba VI. Suppressor activity of bone marrow and spleen cells in C57Bl/6 mice during carcinogenesis induced by 7,12-dimethylbenz(a)anthracene. Eksperimental'naia onkologiia. 1989;11:23–26.
    1. Srivastava MK, et al. Targeting myeloid-derived suppressor cells augments antitumor activity against lung cancer. ImmunoTargets and therapy. 2012;2012:7–12.
    1. Lu T, et al. Tumor-infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice. J Clin Invest. 2011;121:4015–4029. doi: 10.1172/JCI45862.
    1. Sim SH, et al. Influence of chemotherapy on nitric oxide synthase, indole-amine-2,3-dioxygenase and CD124 expression in granulocytes and monocytes of non-small cell lung cancer. Cancer Sci. 2012;103:155–160. doi: 10.1111/j.1349-7006.2011.02158.x.
    1. Srivastava MK, et al. Myeloid suppressor cells and immune modulation in lung cancer. Immunotherapy. 2012;4:291–304. doi: 10.2217/imt.11.178.
    1. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 2009;9:798–809. doi: 10.1038/nrc2734.
    1. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12:253–268. doi: 10.1038/nri3175.
    1. Ostrand-Rosenberg S. Myeloid-derived suppressor cells: more mechanisms for inhibiting antitumor immunity. Cancer Immunol Immunother. 2010;59:1593–1600. doi: 10.1007/s00262-010-0855-8.
    1. Bronte V, Serafini P, Mazzoni A, Segal DM, Zanovello P. L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol. 2003;24:302–306. doi: 10.1016/S1471-4906(03)00132-7.
    1. Wu G, Morris SM., Jr Arginine metabolism: nitric oxide and beyond. Biochem J. 1998;336:1–17. doi: 10.1042/bj3360001.
    1. Nagaraj S, Schrum AG, Cho HI, Celis E, Gabrilovich DI. Mechanism of T cell tolerance induced by myeloid-derived suppressor cells. J Immunol. 2010;184:3106–3116. doi: 10.4049/jimmunol.0902661.
    1. Jiang Y, Li Y, Zhu B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 2015;6:e1792. doi: 10.1038/cddis.2015.162.
    1. Tsukumo SI, Yasutomo K. Regulation of CD8(+) T cells and antitumor immunity by notch signaling. Front Immunol. 2018;9:101. doi: 10.3389/fimmu.2018.00101.
    1. Liu Q, et al. Plasmodium parasite as an effective hepatocellular carcinoma antigen glypican-3 delivery vector. Oncotarget. 2017;8:24785–24796.
    1. Shin SC, Vanderberg JP, Terzakis JA. Direct infection of hepatocytes by sporozoites of Plasmodium berghei. J Protozool. 1982;29:448–454. doi: 10.1111/j.1550-7408.1982.tb05431.x.
    1. Regev-Rudzki N, et al. Cell-cell communication between malaria-infected red blood cells via exosome-like vesicles. Cell. 2013;153:1120–1133. doi: 10.1016/j.cell.2013.04.029.

Source: PubMed

3
Suscribir