Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Lactate modulates CD4+ T-cell polarization and induces an immunosuppressive environment, which sustains prostate carcinoma progression via TLR8/miR21 axis

Abstract

Leukocyte infiltration plays an active role in controlling tumor development. In the early stages of carcinogenesis, T cells counteract tumor growth. However, in advanced stages, cancer cells and infiltrating stromal components interfere with the immune control and instruct immune cells to support, rather than counteract, tumor malignancy, via cell–cell contact or soluble mediators. In particular, metabolites are emerging as active players in driving immunosuppression. Here we demonstrate that in a prostate cancer model lactate released by glycolytic cancer-associated fibroblasts (CAFs) acts on CD4+ T cells, shaping T-cell polarization. In particular, CAFs exposure (i) reduces the percentage of the antitumoral Th1 subset, inducing a lactate-dependent, SIRT1-mediated deacetylation/degradation of T-bet transcription factor; (ii) increases Treg cells, driving naive T cells polarization, through a lactate-based NF-kB activation and FoxP3 expression. In turn, this metabolic-based CAF-immunomodulated environment exerts a pro-invasive effect on prostate cancer cells, by activating a previously unexplored miR21/TLR8 axis that sustains cancer malignancy.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–37.

    Article  CAS  Google Scholar 

  2. Lyssiotis CA, Kimmelman AC. Metabolic interactions in the tumor microenvironment. Trends Cell Biol. 2017;27:863–75.

    Article  CAS  Google Scholar 

  3. Anari F, Ramamurthy C, Zibelman M. Impact of tumor microenvironment composition on therapeutic responses and clinical outcomes in cancer. Future Oncol. 2018;14:1409–21.

    Article  CAS  Google Scholar 

  4. Fiaschi T, Marini A, Giannoni E, Taddei ML, Gandellini P, De Donatis A, et al. Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay. Cancer Res. 2012;72:5130–40.

    Article  CAS  Google Scholar 

  5. Giannoni E, Taddei ML, Morandi A, Comito G, Calvani M, Bianchini F, et al. Targeting stromal-induced pyruvate kinase M2 nuclear translocation impairs oxphos and prostate cancer metastatic spread. Oncotarget. 2015;6:24061–74.

    Article  Google Scholar 

  6. Wang X, Teng F, Kong L, Yu J. PD-L1 expression in human cancers and its association with clinical outcomes. Onco Targets Ther. 2016;9:5023–39.

    Article  CAS  Google Scholar 

  7. Alsaab HO, Sau S, Alzhrani R, Tatiparti K, Bhise K, Kashaw SK, et al. PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol. 2017;8:561.

    Article  Google Scholar 

  8. Ludin A, Zon LI. Cancer immunotherapy: the dark side of PD-1 receptor inhibition. Nature. 2017;552:41–42.

    Article  CAS  Google Scholar 

  9. Macintyre AN, Gerriets VA, Nichols AG, Michalek RD, Rudolph MC, Deoliveira D, et al. The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function. Cell Metab. 2014;20:61–72.

    Article  CAS  Google Scholar 

  10. Biswas SK. Metabolic reprogramming of immune cells in cancer progression. Immunity. 2015;43:435–49.

    Article  CAS  Google Scholar 

  11. Buck MD, Sowell RT, Kaech SM, Pearce EL. Metabolic instruction of immunity. Cell. 2017;169:570–86.

    Article  CAS  Google Scholar 

  12. Sugiura A, Rathmell JC. Metabolic barriers to T cell function in tumors. J Immunol. 2018;200:400–7.

    Article  CAS  Google Scholar 

  13. Angelin A, Gil-de-Gómez L, Dahiya S, Jiao J, Guo L, Levine MH, et al. Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab. 2017;25:1282–93.e1287.

    Article  CAS  Google Scholar 

  14. Geltink RIK, Kyle RL, Pearce EL. Unraveling the complex interplay between T cell metabolism and function. Annu Rev Immunol. 2018;36:461–88.

    Article  CAS  Google Scholar 

  15. Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 2014;513:559–63.

    Article  CAS  Google Scholar 

  16. Fischer K, Hoffmann P, Voelkl S, Meidenbauer N, Ammer J, Edinger M, et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 2007;109:3812–9.

    Article  CAS  Google Scholar 

  17. Husain Z, Huang Y, Seth P, Sukhatme VP. Tumor-derived lactate modifies antitumor immune response: effect on myeloid-derived suppressor cells and NK cells. J Immunol. 2013;191:1486–95.

    Article  CAS  Google Scholar 

  18. Romero-Garcia S, Moreno-Altamirano MM, Prado-Garcia H, Sánchez-García FJ. Lactate contribution to the tumor microenvironment: mechanisms, effects on immune cells and therapeutic relevance. Front Immunol. 2016;7:52.

    Article  Google Scholar 

  19. Huber V, Camisaschi C, Berzi A, Ferro S, Lugini L, Triulzi T, et al. Cancer acidity: an ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol. 2017;43:74–89.

    Article  CAS  Google Scholar 

  20. Morrot A, da Fonseca LM, Salustiano EJ, Gentile LB, Conde L, Filardy AA, et al. Metabolic symbiosis and immunomodulation: how tumor cell-derived lactate may dsturb innate and adaptive immune responses. Front Oncol. 2018;8:81.

    Article  Google Scholar 

  21. Pavlides S, Whitaker-Menezes D, Castello-Cros R, Flomenberg N, Witkiewicz AK, Frank PG, et al. The reverse Warburg effect: aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle. 2009;8:3984–4001.

    Article  CAS  Google Scholar 

  22. Saada A. Mitochondria: mitochondrial OXPHOS (dys) function ex vivo--the use of primary fibroblasts. Int J Biochem Cell Biol. 2014;48:60–65.

    Article  CAS  Google Scholar 

  23. Sotgia F, Whitaker-Menezes D, Martinez-Outschoorn UE, Flomenberg N, Birbe RC, Witkiewicz AK, et al. Mitochondrial metabolism in cancer metastasis: visualizing tumor cell mitochondria and the “reverse Warburg effect” in positive lymph node tissue. Cell Cycle. 2012;11:1445–54.

    Article  CAS  Google Scholar 

  24. Yoshida GJ. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. J Exp Clin Cancer Res. 2015;34:111.

    Article  Google Scholar 

  25. Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016;41:211–8.

    Article  CAS  Google Scholar 

  26. Fiaschi T, Chiarugi P. Oxidative stress, tumor microenvironment, and metabolic reprogramming: a diabolic liaison. Int J Cell Biol. 2012;2012:762825.

    Article  Google Scholar 

  27. Su S, Liao J, Liu J, Huang D, He C, Chen F, et al. Blocking the recruitment of naive CD4. Cell Res. 2017;27:461–82.

    Article  CAS  Google Scholar 

  28. Ziani L, Chouaib S, Thiery J. Alteration of the antitumor immune response by cancer-associated fibroblasts. Front Immunol. 2018;9:414.

    Article  Google Scholar 

  29. Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 2018;33:463–79.e410.

    Article  CAS  Google Scholar 

  30. Johnston CJ, Smyth DJ, Dresser DW, Maizels RM. TGF-β in tolerance, development and regulation of immunity. Cell Immunol. 2016;299:14–22.

    Article  CAS  Google Scholar 

  31. Neuzillet C, Tijeras-Raballand A, Cohen R, Cros J, Faivre S, Raymond E, et al. Targeting the TGFβ pathway for cancer therapy. Pharmacol Ther. 2015;147:22–31.

    Article  CAS  Google Scholar 

  32. Thompson ED, Enriquez HL, Fu YX, Engelhard VH. Tumor masses support naive T cell infiltration, activation, and differentiation into effectors. J Exp Med. 2010;207:1791–804.

    Article  CAS  Google Scholar 

  33. Levine BL, Bernstein WB, Connors M, Craighead N, Lindsten T, Thompson CB, et al. Effects of CD28 costimulation on long-term proliferation of CD4 + T cells in the absence of exogenous feeder cells. J Immunol. 1997;159:5921–30.

    CAS  PubMed  Google Scholar 

  34. Fang D, Zhu J. Dynamic balance between master transcription factors determines the fates and functions of CD4 T cell and innate lymphoid cell subsets. J Exp Med. 2017;214:1861–76.

    Article  CAS  Google Scholar 

  35. Gambini J, Gomez-Cabrera MC, Borras C, Valles SL, Lopez-Grueso R, Martinez-Bello VE, et al. Free [NADH]/[NAD( + )] regulates sirtuin expression. Arch Biochem Biophys. 2011;512:24–29.

    Article  CAS  Google Scholar 

  36. Guarente L. Calorie restriction and sirtuins revisited. Genes Dev. 2013;27:2072–85.

    Article  CAS  Google Scholar 

  37. Houtkooper RH, Pirinen E, Auwerx J. Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Cell Biol. 2012;13:225–38.

    Article  CAS  Google Scholar 

  38. van Loosdregt J, Brunen D, Fleskens V, Pals CE, Lam EW, Coffer PJ. Rapid temporal control of Foxp3 protein degradation by sirtuin-1. PLoS ONE. 2011;6:e19047.

    Article  Google Scholar 

  39. Sonveaux P, Copetti T, De Saedeleer CJ, Végran F, Verrax J, Kennedy KM, et al. Targeting the lactate transporter MCT1 in endothelial cells inhibits lactate-induced HIF-1 activation and tumor angiogenesis. PLoS ONE. 2012;7:e33418.

    Article  CAS  Google Scholar 

  40. Végran F, Boidot R, Michiels C, Sonveaux P, Feron O. Lactate influx through the endothelial cell monocarboxylate transporter MCT1 supports an NF-κB/IL-8 pathway that drives tumor angiogenesis. Cancer Res. 2011;71:2550–60.

    Article  Google Scholar 

  41. Polet F, Feron O. Endothelial cell metabolism and tumour angiogenesis: glucose and glutamine as essential fuels and lactate as the driving force. J Intern Med. 2013;273:156–65.

    Article  CAS  Google Scholar 

  42. Long M, Park SG, Strickland I, Hayden MS, Ghosh S. Nuclear factor-kappaB modulates regulatory T cell development by directly regulating expression of Foxp3 transcription factor. Immunity. 2009;31:921–31.

    Article  CAS  Google Scholar 

  43. Ruan Q, Chen YH. Nuclear factor-κB in immunity and inflammation: the Treg and Th17 connection. Adv Exp Med Biol. 2012;946:207–21.

    Article  CAS  Google Scholar 

  44. Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, et al. MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci USA. 2012;109:E2110–2116.

    Article  CAS  Google Scholar 

  45. Zhang Q, Helfand BT, Jang TL, Zhu LJ, Chen L, Yang XJ, et al. Nuclear factor-kappaB-mediated transforming growth factor-beta-induced expression of vimentin is an independent predictor of biochemical recurrence after radical prostatectomy. Clin Cancer Res. 2009;15:3557–67.

    Article  CAS  Google Scholar 

  46. Giannoni E, Bianchini F, Calorini L, Chiarugi P. Cancer associated fibroblasts exploit reactive oxygen species through a proinflammatory signature leading to epithelial mesenchymal transition and stemness. Antioxid Redox Signal. 2011;14:2361–71.

    Article  CAS  Google Scholar 

  47. Montanari M, Rossetti S, Cavaliere C, D’Aniello C, Malzone MG, Vanacore D, et al. Epithelial-mesenchymal transition in prostate cancer: an overview. Oncotarget. 2017;8:35376–89.

    Article  Google Scholar 

  48. Mendler AN, Hu B, Prinz PU, Kreutz M, Gottfried E, Noessner E. Tumor lactic acidosis suppresses CTL function by inhibition of p38 and JNK/c-Jun activation. Int J Cancer. 2012;131:633–40.

    Article  CAS  Google Scholar 

  49. Renner K, Singer K, Koehl GE, Geissler EK, Peter K, Siska PJ, et al. Metabolic hallmarks of tumor and immune cells in the tumor microenvironment. Front Immunol. 2017;8:248.

    Article  Google Scholar 

  50. Gottfried E, Kunz-Schughart LA, Ebner S, Mueller-Klieser W, Hoves S, Andreesen R, et al. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood. 2006;107:2013–21.

    Article  CAS  Google Scholar 

  51. Shime H, Yabu M, Akazawa T, Kodama K, Matsumoto M, Seya T, et al. Tumor-secreted lactic acid promotes IL-23/IL-17 proinflammatory pathway. J Immunol. 2008;180:7175–83.

    Article  CAS  Google Scholar 

  52. Haas R, Smith J, Rocher-Ros V, Nadkarni S, Montero-Melendez T, D’Acquisto F, et al. Lactate regulates metabolic and pro-inflammatory circuits in control of T cell migration and effector functions. PLoS Biol. 2015;13:e1002202.

    Article  Google Scholar 

  53. Calcinotto A, Filipazzi P, Grioni M, Iero M, De Milito A, Ricupito A, et al. Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Res. 2012;72:2746–56.

    Article  CAS  Google Scholar 

  54. Pearce EL. Metabolism in T cell activation and differentiation. Curr Opin Immunol. 2010;22:314–20.

    Article  CAS  Google Scholar 

  55. Siska PJ, Rathmell JC. T cell metabolic fitness in antitumor immunity. Trends Immunol. 2015;36:257–64.

    Article  CAS  Google Scholar 

  56. MacIver NJ, Michalek RD, Rathmell JC. Metabolic regulation of T lymphocytes. Annu Rev Immunol. 2013;31:259–83.

    Article  CAS  Google Scholar 

  57. Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, et al. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4 + T cell subsets. J Immunol. 2011;186:3299–303.

    Article  CAS  Google Scholar 

  58. Murray CM, Hutchinson R, Bantick JR, Belfield GP, Benjamin AD, Brazma D, et al. Monocarboxylate transporter MCT1 is a target for immunosuppression. Nat Chem Biol. 2005;1:371–6.

    Article  CAS  Google Scholar 

  59. Brand A, Singer K, Koehl GE, Kolitzus M, Schoenhammer G, Thiel A, et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 2016;24:657–71.

    Article  CAS  Google Scholar 

  60. Eidelman E, Twum-Ampofo J, Ansari J, Siddiqui MM. The metabolic phenotype of prostate cancer. Front Oncol. 2017;7:131.

    Article  Google Scholar 

  61. Hensley CT, Wasti AT, DeBerardinis RJ. Glutamine and cancer: cell biology, physiology, and clinical opportunities. J Clin Invest. 2013;123:3678–84.

    Article  CAS  Google Scholar 

  62. Mishra R, Haldar S, Placencio V, Madhav A, Rohena-Rivera K, Agarwal P, et al. Stromal epigenetic alterations drive metabolic and neuroendocrine prostate cancer reprogramming. J Clin Invest. 2018;128:4472–84.

    Article  Google Scholar 

  63. Metzler B, Gfeller P, Guinet E. Restricting glutamine or glutamine-dependent purine and pyrimidine syntheses promotes human T cells with high FOXP3 expression and regulatory properties. J Immunol. 2016;196:3618–30.

    Article  CAS  Google Scholar 

  64. Carr EL, Kelman A, Wu GS, Gopaul R, Senkevitch E, Aghvanyan A, et al. Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J Immunol. 2010;185:1037–44.

    Article  CAS  Google Scholar 

  65. Wang F, Chan CH, Chen K, Guan X, Lin HK, Tong Q. Deacetylation of FOXO3 by SIRT1 or SIRT2 leads to Skp2-mediated FOXO3 ubiquitination and degradation. Oncogene. 2012;31:1546–57.

    Article  CAS  Google Scholar 

  66. Ha TY. The role of regulatory T cells in cancer. Immune Netw. 2009;9:209–35.

    Article  Google Scholar 

  67. Flammiger A, Weisbach L, Huland H, Tennstedt P, Simon R, Minner S, et al. High tissue density of FOXP3 + T cells is associated with clinical outcome in prostate cancer. Eur J Cancer. 2013;49:1273–9.

    Article  CAS  Google Scholar 

  68. Krichevsky AM, Gabriely G. miR-21: a small multi-faceted RNA. J Cell Mol Med. 2009;13:39–53.

    Article  CAS  Google Scholar 

  69. Hatley ME, Patrick DM, Garcia MR, Richardson JA, Bassel-Duby R, van Rooij E, et al. Modulation of K-Ras-dependent lung tumorigenesis by microRNA-21. Cancer Cell. 2010;18:282–93.

    Article  CAS  Google Scholar 

  70. Pfeffer SR, Yang CH, Pfeffer LM. The role of miR-21 in cancer. Drug Dev Res. 2015;76:270–7.

    Article  CAS  Google Scholar 

  71. Lim S, Phillips JB, Madeira da Silva L, Zhou M, Fodstad O, Owen LB, et al. Interplay between immune checkpoint proteins and cellular metabolism. Cancer Res. 2017;77:1245–9.

    Article  CAS  Google Scholar 

  72. Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162:1229–41.

    Article  CAS  Google Scholar 

  73. Hude I, Sasse S, Engert A, Bröckelmann PJ. The emerging role of immune checkpoint inhibition in malignant lymphoma. Haematologica. 2017;102:30–42.

    Article  CAS  Google Scholar 

  74. Giannoni E, Bianchini F, Masieri L, Serni S, Torre E, Calorini L, et al. Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and cancer stemness. Cancer Res. 2010;70:6945–56.

    Article  CAS  Google Scholar 

  75. Wong N, Yan J, Ojo D, De Melo J, Cutz JC, Tang D. Changes in PKM2 associate with prostate cancer progression. Cancer Invest. 2014;32:330–8.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The work was supported by Fondazione Umberto Veronesi to GC and AM, Associazione Italiana Ricerca sul Cancro (AIRC) (grant 8797 to PC), AIRC and Fondazione Cassa di Risparmio di Firenze (grant 19515 to PC and AM), Istituto Toscano Tumori (grant 0203607 to PC), Programma operativo regionale Obiettivo “Competitività regionale e occupazione” della Regione Toscana cofinanziato dal Fondo europeo di sviluppo regionale 2007-2013 (POR CReO FESR 2007-2013, grant to PC). The authors thank Dr. Lavinia Ferrone for technical support.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to E. Giannoni.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Comito, G., Iscaro, A., Bacci, M. et al. Lactate modulates CD4+ T-cell polarization and induces an immunosuppressive environment, which sustains prostate carcinoma progression via TLR8/miR21 axis. Oncogene 38, 3681–3695 (2019). https://doi.org/10.1038/s41388-019-0688-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-019-0688-7

This article is cited by

Search

Quick links