Myeloid-derived suppressor cells modulate immune responses independently of NADPH oxidase in the ovarian tumor microenvironment in mice

Heidi E Godoy, A Nazmul H Khan, R Robert Vethanayagam, Melissa J Grimm, Kelly L Singel, Nonna Kolomeyevskaya, Kevin J Sexton, Anupama Parameswaran, Scott I Abrams, Kunle Odunsi, Brahm H Segal, Heidi E Godoy, A Nazmul H Khan, R Robert Vethanayagam, Melissa J Grimm, Kelly L Singel, Nonna Kolomeyevskaya, Kevin J Sexton, Anupama Parameswaran, Scott I Abrams, Kunle Odunsi, Brahm H Segal

Abstract

The phagocyte NADPH oxidase generates superoxide anion and downstream reactive oxidant intermediates in response to infectious threat, and is a critical mediator of antimicrobial host defense and inflammatory responses. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that are recruited by cancer cells, accumulate locally and systemically in advanced cancer, and can abrogate anti-tumor immunity. Prior studies have implicated the phagocyte NADPH oxidase as being an important component promoting MDSC accumulation and immunosuppression in cancer. We therefore used engineered NADPH oxidase-deficient (p47 (phox-/-)) mice to delineate the role of this enzyme complex in MDSC accumulation and function in a syngeneic mouse model of epithelial ovarian cancer. We found that the presence of NADPH oxidase did not affect tumor progression. The accumulation of MDSCs locally and systemically was similar in tumor-bearing wild-type (WT) and p47 (phox-/-) mice. Although MDSCs from tumor-bearing WT mice had functional NADPH oxidase, the suppressive effect of MDSCs on ex vivo stimulated T cell proliferation was NADPH oxidase-independent. In contrast to other tumor-bearing mouse models, our results show that MDSC accumulation and immunosuppression in syngeneic epithelial ovarian cancer is NADPH oxidase-independent. We speculate that factors inherent to the tumor, tumor microenvironment, or both determine the specific requirement for NADPH oxidase in MDSC accumulation and function.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1.Time. to tumor progression requiring euthanasia…
Figure 1.Time. to tumor progression requiring euthanasia is not altered by NADPH oxidase.
Kaplan-Meier plots of WT and NADPH oxidase-deficient (p47phox−/−) mice (10 mice/group) showed similar survival after i.p. MOSEC challenge (log-rank, p = 0.25).
Figure 2. Effect of NADPH oxidase in…
Figure 2. Effect of NADPH oxidase in local and systemic accumulation of MDSCs in tumor-bearing mice.
A) Representative quantification of MDSCs. Splenocytes from WT and p47phox−/− mice at day 42 and 90 after MOSEC administration were analyzed for MDSC accumulation. Gating on myeloid (CD11b+) cells, the proportion of monocytic MDSCs (R1; Ly6C+Ly6G−) and granulocytic MDSCs (R2; Ly6G+Ly6CLow) significantly increased at day 90 versus day 42. All gates were set based on isotypes. This approach was used to quantify MDSCs in PECs, lymph nodes, and spleens. B) Proportion of MDSCs in myeloid PECs on day 42 and 90. The proportion with granulocytic and monocytic MDSC markers was greater in advanced (day 90) versus early (day 42) stage tumor burden in both genotypes. C) In draining lymph nodes, there was a trend toward increased monocytic MDSC accumulation in p47phox−/− versus WT mice at day 42 but not at day 90. There was no effect of NADPH oxidase on granulocytic MDSC accumulation at either time point. D) In spleens, there was an increased accumulation of MDSCs, particularly granulocytic MDSCs, in mice with advanced versus early disease, but no effect of mouse genotype. Data (± SEM) are from at least 3 mice per genotype per time point, and are representative of 3 separate experiments. Comparison between genotypes: p = NS.
Figure 3. Role of NADPH oxidase in…
Figure 3. Role of NADPH oxidase in cytokine production in the ovarian tumor microenvironment.
Cell-free supernatants collected from ascites from WT and p47phox−/− mice at day 90 after MOSEC administration were analyzed by ELISA for pro-inflammatory cytokines, G-CSF, and VEGF. N = 8 mice per genotype. Comparison between genotypes: p = NS.
Figure 4. The p47 phox component is…
Figure 4. The p47phox component is required for NADPH oxidase activity in granulocytic MDSCs.
PECs from WT and p47phox−/− mice harvested at day 90 after MOSEC challenge were stimulated with PMA, and intracellular ROI production in CD11b+Ly6G+ cells was assessed by H2DCFDA fluorescence. A) Gating on all non-aggregated cells (Gate 1), CD11b+Ly6G+ cells (Gate 2) were defined using respective isotype controls. B and C) Stimulated ROI production was detectable in WT (B), but not in NADPH oxidase-deficient (C), granulocytic MDSCs. White plot = PMA stimulation; shaded plot = no-stimulation. Results are representative of two experiments.
Figure 5. Analysis of peritoneal exudate cells…
Figure 5. Analysis of peritoneal exudate cells from MOSEC-bearing mice after enrichment for myeloid cells and granulocytic cells.
Myeloid cells and granulocytic cells from PECs of MOSEC-bearing WT and p47phox−/− mice were column-purified using anti-CD11b and anti-Ly6G, respectively. Analysis of post-enriched fractions showed concordance between cytology and flow cytometry. Representative analysis of PECs from p47phox−/− mice collected on 90 after MOSEC administration is shown. A) The CD11b-negative fraction contained a preponderance of tumor cells (based on cytology), while myeloid cells were rare. B) The CD11b-enriched fraction contained a mixed myeloid cell population, with a preponderance of macrophages based on cytology and surface markers (CD11b+F4/80+). C) The Ly6G-enriched fraction contained a preponderance of granulocytic cells (arrows), with the majority of myeloid cells expressing granulocytic MDSC markers (CD11b+Ly6G+Ly6Clow).
Figure 6. Myeloid peritoneal cells in MOSEC-bearing…
Figure 6. Myeloid peritoneal cells in MOSEC-bearing mice suppress T cell proliferation independently of NADPH oxidase.
Purified myeloid (CD11b+) PECs from MOSEC-bearing WT and p47phox−/− mice were co-cultured with CFSE-labeled splenocytes from naïve mice (E∶T ratio: 1∶1) in anti-CD3/B7.1-coated plates. After 72 h of culture, CD4+ and CD8+ T cell proliferation was assessed based on CFSE dilution as described in methods. A) Representation histograms showing that CD11b+, but not CD11b−, PECs from MOSEC-bearing WT and p47phox−/− mice (day 90) completely suppress anti-CD3/B7.1-stimulated CD4+ T cells proliferation. Anti-CD3/B7.1 stimulated and unstimulated CD4+ T cells are used as positive and negative controls, respectively. B) CD11b-enriched PECs from MOSEC-bearing WT and p47phox−/− mice at day 42 equally suppress both CD4+ and CD8+ T cell proliferation. In contrast, CD11b− PECs from the same mice incompletely suppressed anti-CD3/B7.1-stimulated T cell proliferation. C) CD11b-enriched PECs from MOSEC-bearing WT and p47phox−/− mice at day 90 completely suppressed T cell proliferation while the CD11b-negative fraction had no significant effect on T cell proliferation. Myeloid PECs collected on day 42 were pooled because of limited cell number, while non-pooled PECs from individual mice were analyzed from day 90 harvests. Individual experiments were performed using PECs from 3 mice per genotype per time point, and each experiment was repeated at least once using PECs from different mice, with similar results. Comparison between genotypes: p = NS.
Figure 7. Peritoneal and splenic granulocytic MDSCs…
Figure 7. Peritoneal and splenic granulocytic MDSCs from tumor-bearing mice suppress T cell proliferation independently of NADPH oxidase.
Ly6G-enriched PECs and splenocytes from MOSEC-bearing WT and p47phox−/− mice (day 90) were co-cultured with splenocytes from non-tumor-bearing WT mice (E∶T ratio: 1∶1). A) Ly6G-enriched PECs completely suppressed anti-CD3/B7.1-stimulated CD4+ and CD8+ T cell proliferation. PECs from 3 mice per genotype were evaluated. B) In Ly6G-enriched splenocytes, the majority of cells had a granulocytic morphology (arrows), and 86% of CD11b+ cells expressed granulocytic MDSC markers (Ly6G+Ly6Clow). C) Ly6G-enriched splenocytes from WT and p47phox−/− mice modestly suppressed anti-CD3/B7.1-stimulated CD4+ and CD8+ T cell proliferation. N = 3 mice per genotype were used in this experiment, and results are representative of 3 experiments. Comparison between genotypes: p = NS.

References

    1. Sinha P, Parker KH, Horn L, Ostrand-Rosenberg S (2012) Tumor-induced myeloid-derived suppressor cell function is independent of IFN-gamma and IL-4Ralpha. Eur J Immunol 42: 2052–2059.
    1. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI (2008) Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol 181: 5791–5802.
    1. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V (2012) Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 12: 253–268.
    1. Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, et al. (2007) Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med 13: 828–835.
    1. Srivastava MK, Zhu L, Harris-White M, Kar U, Huang M, et al. (2012) Myeloid suppressor cell depletion augments antitumor activity in lung cancer. PLoS ONE 7: e40677.
    1. Hanson EM, Clements VK, Sinha P, Ilkovitch D, Ostrand-Rosenberg S (2009) Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cells. J Immunol 183: 937–944.
    1. Sinha P, Okoro C, Foell D, Freeze HH, Ostrand-Rosenberg S, et al. (2008) Proinflammatory S100 proteins regulate the accumulation of myeloid-derived suppressor cells. J Immunol 181: 4666–4675.
    1. Li H, Han Y, Guo Q, Zhang M, Cao X (2009) Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J Immunol 182: 240–249.
    1. Fortin C, Huang X, Yang Y (2012) NK cell response to vaccinia virus is regulated by myeloid-derived suppressor cells. J Immunol 189: 1843–1849.
    1. Ochoa AC, Zea AH, Hernandez C, Rodriguez PC (2007) Arginase, prostaglandins, and myeloid-derived suppressor cells in renal cell carcinoma. Clin Cancer Res 13: 721s–726s.
    1. Highfill SL, Rodriguez PC, Zhou Q, Goetz CA, Koehn BH, et al. (2010) Bone marrow myeloid-derived suppressor cells (MDSCs) inhibit graft-versus-host disease (GVHD) via an arginase-1-dependent mechanism that is up-regulated by interleukin-13. Blood 116: 5738–5747.
    1. Serafini P, Mgebroff S, Noonan K, Borrello I (2008) Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells. Cancer Res 68: 5439–5449.
    1. Pan PY, Ma G, Weber KJ, Ozao-Choy J, Wang G, et al. (2010) 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 70: 99–108.
    1. Kujawski M, Kortylewski M, Lee H, Herrmann A, Kay H, et al. (2008) Stat3 mediates myeloid cell-dependent tumor angiogenesis in mice. J Clin Invest 118: 3367–3377.
    1. Winter WE 3rd, Maxwell GL, Tian C, Carlson JW, Ozols RF, et al. (2007) Prognostic factors for stage III epithelial ovarian cancer: a Gynecologic Oncology Group Study. J Clin Oncol 25: 3621–3627.
    1. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, et al. (2003) Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 348: 203–213.
    1. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, et al. (2005) Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A 102: 18538–18543.
    1. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, et al. (2004) Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 10: 942–949.
    1. Scarlett UK, Rutkowski MR, Rauwerdink AM, Fields J, Escovar-Fadul X, et al. (2012) Ovarian cancer progression is controlled by phenotypic changes in dendritic cells. J Exp Med 209: 495–506.
    1. Obermajer N, Muthuswamy R, Odunsi KO, Edwards R, Kalinski P (2011) PGE2-Dependent CXCL12 Production and CXCR4 Expression Control the Accumulation of Human MDSCs in Ovarian Cancer Environment. Cancer Res
    1. Segal BH, Veys P, Malech H, Cowan MJ (2011) Chronic granulomatous disease: lessons from a rare disorder. Biol Blood Marrow Transplant 17: S123–131.
    1. Cheng P, Corzo CA, Luetteke N, Yu B, Nagaraj S, et al. (2008) Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein. J Exp Med 205: 2235–2249.
    1. Corzo CA, Cotter MJ, Cheng P, Cheng F, Kusmartsev S, et al. (2009) Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol 182: 5693–5701.
    1. Lechner MG, Liebertz DJ, Epstein AL (2010) Characterization of cytokine-induced myeloid-derived suppressor cells from normal human peripheral blood mononuclear cells. J Immunol 185: 2273–2284.
    1. Lechner MG, Megiel C, Russell SM, Bingham B, Arger N, et al. (2011) Functional characterization of human Cd33+ and Cd11b+ myeloid-derived suppressor cell subsets induced from peripheral blood mononuclear cells co-cultured with a diverse set of human tumor cell lines. J Transl Med 9: 90.
    1. Obermajer N, Muthuswamy R, Odunsi K, Edwards RP, Kalinski P (2011) PGE(2)-induced CXCL12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment. Cancer Res 71: 7463–7470.
    1. Eckhorn R, Gail AM, Bruns A, Gabriel A, Al-Shaikhli B, et al. (2004) Different types of signal coupling in the visual cortex related to neural mechanisms of associative processing and perception. IEEE Trans Neural Netw 15: 1039–1052.
    1. Jackson SH, Gallin JI, Holland SM (1995) The p47phox mouse knock-out model of chronic granulomatous disease. J Exp Med 182: 751–758.
    1. Roby KF, Taylor CC, Sweetwood JP, Cheng Y, Pace JL, et al. (2000) Development of a syngeneic mouse model for events related to ovarian cancer. Carcinogenesis 21: 585–591.
    1. Efimova O, Szankasi P, Kelley TW (2011) Ncf1 (p47phox) is essential for direct regulatory T cell mediated suppression of CD4+ effector T cells. PLoS ONE 6: e16013.
    1. Pollock JD, Williams DA, Gifford MA, Li LL, Du X, et al. (1995) Mouse model of X-linked chronic granulomatous disease, an inherited defect in phagocyte superoxide production. Nat Genet 9: 202–209.
    1. Savina A, Jancic C, Hugues S, Guermonprez P, Vargas P, et al. (2006) NOX2 controls phagosomal pH to regulate antigen processing during crosspresentation by dendritic cells. Cell 126: 205–218.
    1. Jancic C, Savina A, Wasmeier C, Tolmachova T, El-Benna J, et al. (2007) Rab27a regulates phagosomal pH and NADPH oxidase recruitment to dendritic cell phagosomes. Nat Cell Biol 9: 367–378.
    1. Mantegazza AR, Savina A, Vermeulen M, Perez L, Geffner J, et al. (2008) NADPH oxidase controls phagosomal pH and antigen cross-presentation in human dendritic cells. Blood
    1. Segal BH, Han W, Bushey JJ, Joo M, Bhatti Z, et al. (2010) NADPH oxidase limits innate immune responses in the lungs in mice. PLoS ONE 5: e9631.
    1. Segal BH, Grimm MJ, Khan AN, Han W, Blackwell TS (2012) Regulation of innate immunity by NADPH oxidase. Free Radic Biol Med
    1. Romani L, Fallarino F, De Luca A, Montagnoli C, D'Angelo C, et al. (2008) Defective tryptophan catabolism underlies inflammation in mouse chronic granulomatous disease. Nature 451: 211–215.
    1. Ushio-Fukai M, Urao N (2009) Novel role of NADPH oxidase in angiogenesis and stem/progenitor cell function. Antioxid Redox Signal 11: 2517–2533.
    1. Ando T, Mimura K, Johansson CC, Hanson MG, Mougiakakos D, et al. (2008) Transduction with the antioxidant enzyme catalase protects human T cells against oxidative stress. J Immunol 181: 8382–8390.
    1. Sinha P, Clements VK, Ostrand-Rosenberg S (2005) Reduction of myeloid-derived suppressor cells and induction of M1 macrophages facilitate the rejection of established metastatic disease. J Immunol 174: 636–645.
    1. Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI (2004) Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol 172: 989–999.
    1. Kusmartsev S, Gabrilovich DI (2003) Inhibition of myeloid cell differentiation in cancer: the role of reactive oxygen species. J Leukoc Biol 74: 186–196.
    1. Morgenstern DE, Gifford MA, Li LL, Doerschuk CM, Dinauer MC (1997) Absence of respiratory burst in X-linked chronic granulomatous disease mice leads to abnormalities in both host defense and inflammatory response to Aspergillus fumigatus. J Exp Med 185: 207–218.
    1. Chang YC, Segal BH, Holland SM, Miller GF, Kwon-Chung KJ (1998) Virulence of catalase-deficient aspergillus nidulans in p47(phox)-/- mice. Implications for fungal pathogenicity and host defense in chronic granulomatous disease. J Clin Invest 101: 1843–1850.
    1. Meyer C, Sevko A, Ramacher M, Bazhin AV, Falk CS, et al. (2011) Chronic inflammation promotes myeloid-derived suppressor cell activation blocking antitumor immunity in transgenic mouse melanoma model. Proc Natl Acad Sci U S A 108: 17111–17116.
    1. Waight JD, Hu Q, Miller A, Liu S, Abrams SI (2011) Tumor-derived G-CSF facilitates neoplastic growth through a granulocytic myeloid-derived suppressor cell-dependent mechanism. PLoS ONE 6: e27690.
    1. Abrams SI, Waight JD (2012) Identification of a G-CSF-Granulocytic MDSC axis that promotes tumor progression. Oncoimmunology 1: 550–551.
    1. Eruslanov E, Daurkin I, Ortiz J, Vieweg J, Kusmartsev S (2010) Pivotal Advance: Tumor-mediated induction of myeloid-derived suppressor cells and M2-polarized macrophages by altering intracellular PGE(2) catabolism in myeloid cells. J Leukoc Biol 88: 839–848.
    1. Smith C, Chang MY, Parker KH, Beury DW, Duhadaway JB, et al. (2012) IDO Is a Nodal Pathogenic Driver of Lung Cancer and Metastasis Development. Cancer Discov 2: 722–735.
    1. Segal BH, Sakamoto N, Patel M, Maemura K, Klein AS, et al. (2000) Xanthine oxidase contributes to host defense against Burkholderia cepacia in the p47(phox-/-) mouse model of chronic granulomatous disease. Infect Immun 68: 2374–2378.
    1. West AP, Brodsky IE, Rahner C, Woo DK, Erdjument-Bromage H, et al. (2011) TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature 472: 476–480.
    1. Kubo H, Morgenstern D, Quinian WM, Ward PA, Dinauer MC, et al. (1996) Preservation of complement-induced lung injury in mice with deficiency of NADPH oxidase. J Clin Invest 97: 2680–2684.

Source: PubMed

3
Předplatit