Glycolysis regulates the expansion of myeloid-derived suppressor cells in tumor-bearing hosts through prevention of ROS-mediated apoptosis

Shiou-Ling Jian, Wei-Wei Chen, Yu-Chia Su, Yu-Wen Su, Tsung-Hsien Chuang, Shu-Ching Hsu, Li-Rung Huang, Shiou-Ling Jian, Wei-Wei Chen, Yu-Chia Su, Yu-Wen Su, Tsung-Hsien Chuang, Shu-Ching Hsu, Li-Rung Huang

Abstract

Immunotherapy aiming to rescue or boost antitumor immunity is an emerging strategy for treatment of cancers. The efficacy of immunotherapy is strongly controlled by the immunological milieu of cancer patients. Myeloid-derived suppressor cells (MDSCs) are heterogeneous immature myeloid cell populations with immunosuppressive functions accumulating in individuals during tumor progression. The signaling mechanisms of MDSC activation have been well studied. However, there is little known about the metabolic status of MDSCs and the physiological role of their metabolic reprogramming. In this study, we discovered that myeloid cells upregulated their glycolytic genes when encountered with tumor-derived factors. MDSCs exhibited higher glycolytic rate than their normal cell compartment did, which contributed to the accumulation of the MDSCs in tumor-bearing hosts. Upregulation of glycolysis prevented excess reactive oxygen species (ROS) production by MDSCs, which protected MDSCs from apoptosis. Most importantly, we identified the glycolytic metabolite, phosphoenolpyruvate (PEP), as a vital antioxidant agent able to prevent excess ROS production and therefore contributed to the survival of MDSCs. These findings suggest that glycolytic metabolites have important roles in the modulation of fitness of MDSCs and could be potential targets for anti-MDSC strategy. Targeting MDSCs with analogs of specific glycolytic metabolites, for example, 2-phosphoglycerate or PEP may diminish the accumulation of MDSCs and reverse the immunosuppressive milieu in tumor-bearing individuals.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
MDSC accumulation during tumor progression. (a) The IVIS images of Balb/c mice receiving 4T1-LG at indicated time points after implantation. Arrow indicated the IVIS image of the 4T1 lung metastasis in mice. (b) The tumor growth curve and total flux of luciferase activity of mice in (a) (n=3 per group). (c) The hematoxylin and eosin (H&E) staining of lung and liver sections of mice at indicated time points after inoculation of 4T1-LG tumor cells. (d) Representative flow cytometric analysis of Ly6G+ and Ly6C+ populations among gated CD11b+ cells in indicated tissues of control and 4T1-tumor-bearing mice at the third week and sixth week after tumor inoculation. (e) Absolute numbers of total CD11b+ cells, Ly6G+CD11b+ cells and Ly6C+CD11b+ cells in indicated organs (n=3 per group). (f) Immunohistochemical staining for detection of Gr-1+ and CD11b+ cells, respectively, in the liver and tumor sites of 4T1-tumor-bearing mice at indicated time points after tumor inoculation. All the data are representative of two independent experiments. ND, not done; ns, not significant. *P<0.05, **P<0.01, ***P<0.001 (unpaired Student’s t-test). (c and f) Scale bars, 100 μm. (b and e) Error bars, S.D.
Figure 2
Figure 2
Metabolic status in myeloid cells during tumor progression. (a) Heat map summarizing the gene expression data from cDNA microarray. mRNA levels of genes of interest in splenic neutrophils, monocytes from normal healthy mice, gMDSCs and mMDSCs from tumor site of 4T1-tumor-bearing mice were shown. (b) Quantitative real-time PCR analysis of gene expression of glycolytic enzymes in FACSorted CD11b+Ly6C+ and CD11b+Ly6G+ cells from tumor sites of 4T1-tumor-bearing mice and spleens from normal BALB/c mice (n=3 per group). (c) Quantitative real-time PCR analysis of ARG1, NOS2, PDCD1LG1 and PDCD1LG2 in FACSorted CD11b+Ly6C+ and CD11b+Ly6G+ cells from tumor sites of 4T1-tumor-bearing mice and spleens from normal BALB/c mice (n=3 per group). (d) ECAR of FACSorted splenic gMDSCs from 4T1-tumor-bearing mice and splenic neutrophils from normal BALB/c mice (left panel); calculated non-glycolytic acidification, glycolysis and glycolytic capacity of the cells (middle panel); ECAR versus OCR of the above cells under basal condition and oligomycin treatment (right panel, n=7 per group). All the data are representative of two independent experiments. *P<0.05, **P<0.01 and ***P<0.001 (unpaired Student’s t-test). (bd) Error bars, S.D.
Figure 3
Figure 3
In vitro induction of MDSCs. (a) Quantitative real-time PCR analysis of gene expression of glycolytic enzymes in purified bone marrow neutrophils and monocytes after incubation with 4T1-tumor cells or with primary MECs for 40 h. (b) Representative flow cytometric zebra plots of MDSCs induced from bone marrow cells in the presence of GM-CSF for 5 days. CD11b and CD11c expression (left) of the total cells and Ly6C and Ly6G expression (right) of gated CD11b+CD11c− cells were shown. (c) Quantitative real-time PCR analysis of gene expression of glycolytic enzymes in FACSorted GM-CSF-induced (GM-MDSCs) or bone marrow (primary BM) CD11b+CD11c−Ly6C+ and CD11b+CD11c−Ly6G+ cells (n=3 per group). (d) Quantitative real-time PCR analysis of ARG1, NOS2, PDCD1LG1 and PDCD1LG2 in the cells in (c) (n=3 per group). (e) Proliferation of CD4+ (left) and CD8+ (right) T cells cocultured with different numbers of MACSorted fresh bone marrow CD11b+ (BM ctrl) or MACSorted GM-CSF-induced CD11b+ MDSCs (GM-MDSCs). The percentage of the proliferating CD4+ (left panel) and CD8+ (right panel) T cells was calculated by dividing the number of EdU+ cells of each well by the average number of EdU+ cells in wells without MDSCs (as 100%). All the data are representative of two independent experiments. *P<0.05, **P<0.01 and ***P<0.001 (unpaired Student’s t-test). (a and ce) Error bars, S.D.
Figure 4
Figure 4
The in vivo accumulation of MDSCs was dependent on glycolysis. (a) Counts of CD11b+ cells in the blood of PBS-treated (ctrl) or 2-DG-treated normal (N) and 4T1-tumor-bearing (TB) mice at indicated time points after tumor inoculation. Arrow indicated the time point starting 2-DG treatment (n=3 per group). (b) Absolute numbers of CD11b+ cells in the indicated tissues of the mice in (a) at the fourth week after tumor inoculation (n=3 per group). (c) Size of tumor in the PBS-treated (ctrl) and 2-DG-treated tumor-bearing mice measured at the indicated time points (n=6–7 per group). (d) Immunohistochemical staining for detection and (e) quantification of Gr-1+ cells in the tumor and the liver of the PBS-treated (ctrl) and 2-DG-treated tumor-bearing mice at week 4 after tumor inoculation. Arrowheads indicated Gr-1+ cells (n=3 per group). Scale bars, 100 μm. All the data are representative of two independent experiments. ND, not done; ns, not significant. *P<0.05, **P<0.01 and ***P<0.001 (unpaired Student’s t-test). (ac and e) Error bars, S.D.
Figure 5
Figure 5
The expansion of MDSCs was dependent on glycolysis. (a) Percentage of BrdU+ cells among mMDSCs and gMDSCs from the bone marrow (BM), spleen (Sp) and tumor site of the PBS-treated (ctrl) and 2-DG-treated 4T1-tumor-bearing mice (n=3 per group). (b) Proliferation of CD8+ T cells cocultured with different numbers of splenic PBS (ctrl)-, 2-DG-treated CD11b+ MDSCs from 4T1-tumor-bearing mice. (c) Proliferation of CD8+ T cells cocultured with different numbers of splenic PBS (ctrl)-, IA-treated CD11b+ MDSCs from 4T1-tumor-bearing mice. (d) Cell numbers of CD11b+Gr-1+ MDSCs recovered from the MDSC induction culture 5 days after induction using 20 ng/ml of GM-CSF in the presence or absence of 2-DG in 24-well plates. (e) Cell numbers of CD11b+Gr-1+ MDSCs recovered from the MDSC induction culture 3 days after induction using 20 ng/ml of GM-CSF in the presence or absence of 0.5 mM 2-DG and PEP (5 mM) in 96-well plates. (f) Percentage of proliferating CD11b+Gr-1+ MDSCs among the cells from (e). All the data are representative of two independent experiments. ns, not significant. *P<0.05, **P<0.01 and ***P<0.001 (unpaired Student’s t-test). (af) Error bars, S.D.
Figure 6
Figure 6
Glycolysis prevented apoptosis and excess ROS production of MDSCs. (a) Percentage of PI−Annexin V+ early apoptotic cells among mMDSCs and gMDSCs from the bone marrow and spleen of the PBS-treated (ctrl) and 2-DG-treated 4T1-tumor-bearing mice. 4T1-tumor-bearing mice were treated with 40 mg of 2-DG or PBS (ctrl) as described in Materials and Methods (n=3 per group). (b) Representative flow cytometric zebra plots gating on Gr-1+ population of in vitro 4T1-induced myeloid cells subjected to 2-DG or control treatment for 18 or 48 h showed PI and Annexin V staining. The bar graph of percentage of early (PI−Annexin V+) and late (PI+ Annexin V+) apoptotic cells among Gr-1+ population of in vitro 4T1-induced myeloid cells subjected to 2-DG or PBS (ctrl) treatment for 18 or 48 h was shown (n=3 per group). (c) Histogram (left) and bar graph (right) showing fluorescence intensity or mean fluorescence intensity (MFI) of FITC-dUTP-labeled or -unlabeled DNA fragments in MACSorted CD11b+Gr-1+ GM-CSF-induced MDSCs treated with 2-DG or PBS (ctrl) for 22 h and subjected to detection of DNA fragment by TUNEL assay (n=3 per group). (d) ROS production in freshly isolated neutrophils from normal BALB/c mice and in freshly isolated gMDSCs from 4T1-tumor-bearing mice. (e) Representative histograms (left two panels) of ROS production in neutrophils from normal BALB/c mice and in gMDSCs from 4T1-tumor-bearing mice treated with 2-DG or vehicle (DPBS, ctrl) for 2 h. Bar graph (right panel) showing the fold changes of MFI of DCFDA of the cells after treatment with 2-DG for 2 h. The MFI of DCFDA of the cells treated with vehicle was used as denominator (n=3 per group). (f) A representative histogram for ROS production in MACSorted GM-CSF-induced CD11b+Gr-1+ MDSCs treated with or without 10 mM of 2-DG for 6 h. All the data are representative of two independent experiments. ns, not significant. *P<0.05, **P<0.01 and ***P<0.001 (unpaired Student’s t-test). (a–c and e) Error bars, S.D.
Figure 7
Figure 7
PEP ameliorated apoptosis in MDSCs through prevention of excess ROS production during blockade of glycolysis. (a) Percentage of PI−Annexin V+ early apoptotic cells among CD11b+Gr-1+ GM-CSF-induced MDSCs treated with 10 mM of 2-DG or vehicle (ctrl) for 8 h in the presence or absence of PEP at the concentration of 5 mM (n=3 per group). (b) MFI of FITC-dUTP-labeled DNA fragments in MACSorted Gr-1+ GM-CSF-induced MDSCs treated with 10 mM of 2-DG or vehicle (ctrl) for 22 h in the presence or absence of PEP at the concentration of 7.5 mM (n=3 per group). (c) ROS production in CD11b+Gr-1+ GM-CSF-induced MDSCs treated with 10 mM of 2-DG or vehicle (ctrl) for 5 h in the presence or absence of PEP at the concentration of 5 mM (n=3 per group). (d) ROS production in CD11b+Gr-1+ GM-CSF-induced MDSCs pretreated with 2 μM of DPI or vehicle for 1 h before the addition of 2-DG at the concentration of 10 mM for additional incubation for 5 h (n=3 per group). (e) Percentage of PI−Annexin V+ early apoptotic cells among CD11b+Gr-1+ GM-CSF-induced MDSCs treated as in (d) (n=3 per group). All the data are representative of two independent experiments. ns, not significant. *P<0.05, **P<0.01 and ***P<0.001 (unpaired Student’s t-test). (a–e) Error bars, S.D.

References

    1. Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, Kefford R et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet 2014; 384: 1109–1117.
    1. Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 2014; 32: 1020–1030.
    1. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009; 9: 162–174.
    1. Iclozan C, Antonia S, Chiappori A, Chen DT, Gabrilovich D. Therapeutic regulation of myeloid-derived suppressor cells and immune response to cancer vaccine in patients with extensive stage small cell lung cancer. Cancer Immunol Immunother 2013; 62: 909–918.
    1. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012; 12: 253–268.
    1. Kusmartsev S, Cheng F, Yu B, Nefedova Y, Sotomayor E, Lush R et al. All-trans-retinoic acid eliminates immature myeloid cells from tumor-bearing mice and improves the effect of vaccination. Cancer Res 2003; 63: 4441–4449.
    1. Zoglmeier C, Bauer H, Norenberg D, Wedekind G, Bittner P, Sandholzer N et al. CpG blocks immunosuppression by myeloid-derived suppressor cells in tumor-bearing mice. Clin Cancer Res 2011; 17: 1765–1775.
    1. Pearce EL, Pearce EJ. Metabolic pathways in immune cell activation and quiescence. Immunity 2013; 38: 633–643.
    1. Kominsky DJ, Campbell EL, Colgan SP. Metabolic shifts in immunity and inflammation. J Immunol 2010; 184: 4062–4068.
    1. Borregaard N, Herlin T. Energy metabolism of human neutrophils during phagocytosis. J Clin Invest 1982; 70: 550–557.
    1. Krawczyk CM, Holowka T, Sun J, Blagih J, Amiel E, DeBerardinis RJ et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood 2010; 115: 4742–4749.
    1. Kelly B, O'Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res 2015; 25: 771–784.
    1. Fox CJ, Hammerman PS, Thompson CB. Fuel feeds function: energy metabolism and the T-cell response. Nat Rev Immunol 2005; 5: 844–852.
    1. Jones RG, Thompson CB. Revving the engine: signal transduction fuels T cell activation. Immunity 2007; 27: 173–178.
    1. Chang CH, Curtis JD, Maggi LBJr., Faubert B, Villarino AV, O'Sullivan D et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 2013; 153: 1239–1251.
    1. Markowitz J, Wesolowski R, Papenfuss T, Brooks TR, Carson WEIII. Myeloid-derived suppressor cells in breast cancer. Breast Cancer Res Treat 2013; 140: 13–21.
    1. Lechner MG, Liebertz DJ, Epstein AL. Characterization of cytokine-induced myeloid-derived suppressor cells from normal human peripheral blood mononuclear cells. J Immunol 2010; 185: 2273–2284.
    1. Zhao X, Rong L, Zhao X, Li X, Liu X, Deng J et al. TNF signaling drives myeloid-derived suppressor cell accumulation. J Clin Invest 2012; 122: 4094–4104.
    1. Young MRI, Wright MA, Lozano Y, Prechel MM, Benefield J, Leonetti JP et al. Increased recurrence and metastasis in patients whose primary head and neck squamous cell carcinomas secreted granulocyte–macrophage colony-stimulating factor and contained CD34(+) natural suppressor cells. Int J Cancer 1997; 74: 69–74.
    1. Dehqanzada ZA, Storrer CE, Hueman MT, Foley RJ, Harris KA, Jama YH et al. Assessing serum cytokine profiles in breast cancer patients receiving a HER2/neu vaccine using Luminex (R) technology. Oncol Rep 2007; 17: 687–694.
    1. Ghirelli C, Reyal F, Jeanmougin M, Zollinger R, Sirven P, Michea P et al. Breast cancer cell-derived GM-CSF licenses regulatory Th2 induction by plasmacytoid predendritic cells in aggressive disease subtypes. Cancer Res 2015; 75: 2775–2787.
    1. Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Hau CS et al. IL-17-producing gammadelta T cells and neutrophils conspire to promote breast cancer metastasis. Nature 2015; 522: 345–348.
    1. Chandra J, Samali A, Orrenius S. Triggering and modulation of apoptosis by oxidative stress. Free Radic Biol Med 2000; 29: 323–333.
    1. Geering B, Simon HU. Peculiarities of cell death mechanisms in neutrophils. Cell Death Differ 2011; 18: 1457–1469.
    1. Kuijpers TW, Maianski NA, Tool AT, Smit GP, Rake JP, Roos D et al. Apoptotic neutrophils in the circulation of patients with glycogen storage disease type 1b (GSD1b). Blood 2003; 101: 5021–5024.
    1. Kim SY, Jun HS, Mead PA, Mansfield BC, Chou JY. Neutrophil stress and apoptosis underlie myeloid dysfunction in glycogen storage disease type Ib. Blood 2008; 111: 5704–5711.
    1. Boztug K, Appaswamy G, Ashikov A, Schaffer AA, Salzer U, Diestelhorst J et al. A syndrome with congenital neutropenia and mutations in G6PC3. N Engl J Med 2009; 360: 32–43.
    1. Jun HS, Lee YM, Cheung YY, McDermott DH, Murphy PM, De Ravin SS et al. Lack of glucose recycling between endoplasmic reticulum and cytoplasm underlies cellular dysfunction in glucose-6-phosphatase-beta-deficient neutrophils in a congenital neutropenia syndrome. Blood 2010; 116: 2783–2792.
    1. Renshaw SA, Timmons SJ, Eaton V, Usher LR, Akil M, Bingle CD et al. Inflammatory neutrophils retain susceptibility to apoptosis mediated via the Fas death receptor. J Leukocyte Biol 2000; 67: 662–668.
    1. Sinha P, Chornoguz O, Clements VK, Artemenko KA, Zubarev RA, Ostrand-Rosenberg S. Myeloid-derived suppressor cells express the death receptor Fas and apoptose in response to T cell-expressed FasL. Blood 2011; 117: 5381–5390.
    1. Condamine T, Kumar V, Ramachandran IR, Youn JI, Celis E, Finnberg N et al. ER stress regulates myeloid-derived suppressor cell fate through TRAIL-R-mediated apoptosis. J Clin Invest 2014; 124: 2626–2639.
    1. Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol 2004; 4: 181–189.
    1. Corzo CA, Cotter MJ, Cheng P, Cheng F, Kusmartsev S, Sotomayor E et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol 2009; 182: 5693–5701.
    1. Simon HU, Haj-Yehia A, Levi-Schaffer F. Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis 2000; 5: 415–418.
    1. Scheel-Toellner D, Wang K, Craddock R, Webb PR, McGettrick HM, Assi LK et al. Reactive oxygen species limit neutrophil life span by activating death receptor signaling. Blood 2004; 104: 2557–2564.
    1. Kondo Y, Ishitsuka Y, Kadowaki D, Fukumoto Y, Miyamoto Y, Irikura M et al. Phosphoenolpyruvate, a glycolytic intermediate, as a cytoprotectant and antioxidant in ex vivo cold-preserved mouse liver: a potential application for organ preservation. J Pharm Pharmacol 2013; 65: 390–401.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods 2001; 25: 402–408.
    1. Smalley MJ. Isolation, culture and analysis of mouse mammary epithelial cells. Methods Mol Biol 2010; 633: 139–170.

Source: PubMed

3
S'abonner