Skip to main content
Log in

Lactate Exposure Promotes Immunosuppressive Phenotypes in Innate Immune Cells

  • 2020 CMBE Young Innovators issue
  • Published:
Cellular and Molecular Bioengineering Aims and scope Submit manuscript

Abstract

Introduction

Lactate secreted by tumors is not just a byproduct, but rather an active modulator of immune cells. There are few studies aimed at investigating the true effect of lactate, which is normally confounded by pH. Such a knowledge gap needs to be addressed. Herein, we studied the immunomodulatory effects of lactate on dendritic cells (DCs) and macrophages (MΦs).

Methods

Bone marrow-derived innate immune cells were treated with 50 mM sodium lactate (sLA) and incubated for 2 days or 5 days at 37 °C. Controls included media, lipopolysaccharide (LPS), MCT inhibitors (α-cyano-4-hydroxycinnamic acid and AR-C15585). Flow cytometric analysis of immune phenotypes were performed by incubating cells with specific marker antibodies and viability dye. Differential expression analyses were conducted on R using limma-voom and adjusted p-values were generated using the Bejamini-Hochberg Procedure.

Results

Lactate exposure attenuated DC maturation through the downregulation of CD80 and MHCII expression under LPS stimulation. For MΦs, lactate exposure resulted in M2 polarization as evidenced by the reduction of M1 markers (CD38 and iNOS), and the increase in expression of CD163 and Arg1. We also revealed the role of monocarboxylate transporters (MCTs) in mediating lactate effect in MΦs. MCT4 inhibition significantly boosted lactate M2 polarization, while blocking of MCT1/2 failed to reverse the immunosuppressive effect of lactate, correlating with the result of gene expression that lactate increased MCT4 expression, but downregulated the expression of MCT1/2.

Conclusions

This research provides valuable insight on the influence of metabolic products on tumor immunity and will help to identify novel metabolic targets for augmenting cancer immunotherapies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7

Similar content being viewed by others

References

  1. Allen, R. P., A. Bolandparvaz, J. A. Ma, V. A. Manickam, and J. S. Lewis. Latent, immunosuppressive nature of poly(lactic-co-glycolic acid) microparticles. Acs Biomater. Sci. Eng. 4:900–918, 2018. https://doi.org/10.1021/acsbiomaterials.7b00831.

    Article  Google Scholar 

  2. Amici, S. A., et al. CD38 is robustly induced in human macrophages and monocytes in inflammatory conditions. Front. Immunol. 9:1593, 2018.

    Google Scholar 

  3. Annesley, C. E., C. Summers, F. Ceppi, and R. A. Gardner. The evolution and future of CAR T cells for B-cell acute lymphoblastic leukemia. Clin. Pharmacol. Ther. 103:591–598, 2018. https://doi.org/10.1002/cpt.950.

    Article  Google Scholar 

  4. Arlauckas, S. P., et al. Arg1 expression defines immunosuppressive subsets of tumor-associated macrophages. Theranostics 8:5842, 2018.

    Google Scholar 

  5. Banchereau, J. et al. Immunobiology of dendritic cells. Annu. Rev. Immunol. 18, 767- + , https://doi.org/10.1146/annurev.immunol.18.1.767 (2000).

  6. Banchereau, J., and R. M. Steinman. Dendritic cells and the control of immunity. Nature 392:245–252, 1998. https://doi.org/10.1038/32588.

    Article  Google Scholar 

  7. Bosshart, P. D., D. Kalbermatter, S. Bonetti, and D. Fotiadis. Mechanistic basis of L-lactate transport in the SLC16 solute carrier family. Nat. Commun. 10:1–11, 2019.

    Google Scholar 

  8. Brand, A., et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 24:657–671, 2016. https://doi.org/10.1016/j.cmet.2016.08.011.

    Article  Google Scholar 

  9. Bronte, V. Macrophage response to lactic acid Tumor cells hijack macrophages via lactic acid. Immunol. Cell Biol. 92:647–649, 2014. https://doi.org/10.1038/icb.2014.67.

    Article  Google Scholar 

  10. Chen, P., et al. Gpr132 sensing of lactate mediates tumor–macrophage interplay to promote breast cancer metastasis. Proc. Natl. Acad. Sci. U.S.A. 114:580–585, 2017.

    Google Scholar 

  11. Chi, X. W., et al. Significantly increased anti-tumor activity of carcinoembryonic antigen-specific chimeric antigen receptor T cells in combination with recombinant human IL-12. Cancer Med. 8:4753–4765, 2019. https://doi.org/10.1002/cam4.2361.

    Article  Google Scholar 

  12. Colegio, O. R. et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 513, 559– + , https://doi.org/10.1038/nature13490 (2014).

  13. Colen, C. B., et al. Metabolic targeting of lactate efflux by malignant glioma inhibits invasiveness and induces necrosis: an in vivo study. Neoplasia 13:620–632, 2011.

    Google Scholar 

  14. Corbet, C., and O. Feron. Tumour acidosis: from the passenger to the driver’s seat. Nat. Rev. Cancer 17:577–593, 2017. https://doi.org/10.1038/nrc.2017.77.

    Article  Google Scholar 

  15. Crespo, H. J., J. T. Lau, and P. A. Videira. Dendritic cells: a spot on sialic acid. Front. Immunol. 4:491, 2013.

    Google Scholar 

  16. Deng, R., et al. Dimethyl sulfoxide suppresses mouse 4T1 breast cancer growth by modulating tumor-associated macrophage differentiation. J. Breast Cancer 17:25–32, 2014.

    Google Scholar 

  17. Dienel, G. A. Brain lactate metabolism: the discoveries and the controversies. J. Cereb. Blood Flow Metab. 32:1107–1138, 2012.

    Google Scholar 

  18. Elghetany, M. T. Surface marker abnormalities in myelodysplastic syndromes. Haematologica 83:1104–1115, 1998.

    Google Scholar 

  19. Elhelu, M. A. The role of macrophages in immunology. J. Natl Med. Assoc. 75:314–317, 1983.

    Google Scholar 

  20. Enerson, B. E., and L. R. Drewes. Molecular features, regulation, and function of monocarboxylate transporters: implications for drug delivery. J. Pharm. Sci. 92:1531–1544, 2003.

    Google Scholar 

  21. Errea, A. et al. Lactate inhibits the pro-inflammatory response and metabolic reprogramming in murine macrophages in a GPR81-independent manner. PLoS One 11 (2016).

  22. Fischer, K., et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 109:3812–3819, 2007. https://doi.org/10.1182/blood-2006-07-035972.

    Article  Google Scholar 

  23. Förster, R., A. C. Davalos-Misslitz, and A. Rot. CCR7 and its ligands: balancing immunity and tolerance. Nat. Rev. Immunol. 8:362–371, 2008.

    Google Scholar 

  24. Förster, R., et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell 99:23–33, 1999.

    Google Scholar 

  25. Gabrilovich, D. I., S. Ostrand-Rosenberg, and V. Bronte. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 12:253–268, 2012.

    Google Scholar 

  26. Genard, G., S. Lucas, and C. Michiels. Reprogramming of tumor-associated macrophages with anticancer therapies: radiotherapy versus chemo-and immunotherapies. Front. Immunol. 8:828, 2017.

    Google Scholar 

  27. Gottfried, E., et al. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood 107:2013–2021, 2006. https://doi.org/10.1182/blood-2005-05-1795.

    Article  Google Scholar 

  28. Gunn, M. D., et al. Mice lacking expression of secondary lymphoid organ chemokine have defects in lymphocyte homing and dendritic cell localization. J. Exp. Med. 189:451–460, 1999.

    Google Scholar 

  29. Haas, R. et al. Lactate regulates metabolic and pro-inflammatory circuits in control of T cell migration and effector functions. PLoS Biol. 13 (2015).

  30. Hamza, T., J. B. Barnett, and B. Li. Interleukin 12 a key immunoregulatory cytokine in infection applications. Int. J. Mol. Sci. 11:789–806, 2010.

    Google Scholar 

  31. He, Y. T., Q.-M. Zhang, Q. C. Kou, and B. Tang. In vitro generation of cytotoxic T lymphocyte response using dendritic cell immunotherapy in osteosarcoma. Oncol. Lett. 12:1101–1106, 2016.

    Google Scholar 

  32. Hintzen, G., et al. Induction of tolerance to innocuous inhaled antigen relies on a CCR7-dependent dendritic cell-mediated antigen transport to the bronchial lymph node. J. Immunol. 177:7346–7354, 2006.

    Google Scholar 

  33. Hou, B., Y. Tang, W. H. Li, Q. N. Zeng, and D. M. Chang. Efficiency of CAR-T therapy for treatment of solid tumor in clinical trials: a meta-analysis. Dis. Markers 2019. https://doi.org/10.1155/2019/3425291.

    Article  Google Scholar 

  34. Hwang, M. T. P., R. J. Fecek, T. Y. Qin, W. J. Storkus, and Y. D. Wang. Single injection of IL-12 coacervate as an effective therapy against B16-F10 melanoma in mice. J. Control. Release 318:270–278, 2020. https://doi.org/10.1016/j.jconrel.2019.12.035.

    Article  Google Scholar 

  35. Jablonski, K. A. et al. Novel markers to delineate murine M1 and M2 macrophages. PLoS One 10 (2015).

  36. Jang, M. H., et al. CCR7 is critically important for migration of dendritic cells in intestinal lamina propria to mesenteric lymph nodes. J. Immunol. 176:803–810, 2006.

    Google Scholar 

  37. Jin, P., et al. Molecular signatures of maturing dendritic cells: implications for testing the quality of dendritic cell therapies. J. Transl. Med. 8:4, 2010.

    Google Scholar 

  38. Kim, J., and D. J. Mooney. In vivo modulation of dendritic cells by engineered materials: towards new cancer vaccines. Nano today 6:466–477, 2011.

    Google Scholar 

  39. Kiseleva, Y. Y., Shishkin, A. M., Ivanov, A. V., Kulinich, T. M. & Bozhenko, V. K. Car T-cell therapy of solid tumors: promising approaches to modulating antitumor activity of car T cells. Bulletin of Russian State Medical University, 5-12, https://doi.org/10.24075/brsmu.2019.066 (2019).

  40. Kreutz, M., et al. Tumor-derived lactic acid modulates dendritic cell activation and differentiation. Blood 104:147B, 2004.

    Google Scholar 

  41. Lam, J. H., et al. Expression of CD38 on macrophages predicts improved prognosis in hepatocellular carcinoma. Front. Immunol. 10:2093, 2019.

    Google Scholar 

  42. Lanier, L., et al. Correlation of functional properties of human lymphoid cell subsets and surface marker phenotypes using multiparameter analysis and flow cytometry. Immunol. Rev. 74:143–160, 1983.

    Google Scholar 

  43. Lewis, J. S., T. D. Zaveri, C. P. Crooks, II, and B. G. Keselowsky. Microparticle surface modifications targeting dendritic cells for non-activating applications. Biomaterials 33:7221–7232, 2012. https://doi.org/10.1016/j.biomaterials.2012.06.049.

    Article  Google Scholar 

  44. Martín-Fontecha, A., et al. Regulation of dendritic cell migration to the draining lymph node: impact on T lymphocyte traffic and priming. J. Exp. Med. 198:615–621, 2003.

    Google Scholar 

  45. Mendler, A. N., et al. Tumor lactic acidosis suppresses CTL function by inhibition of p38 and JNK/c-Jun activation. Int. J. Cancer 131:633–640, 2012. https://doi.org/10.1002/ijc.26410.

    Article  Google Scholar 

  46. Mills, C. D., L. L. Lenz, and R. A. Harris. A breakthrough: macrophage-directed cancer immunotherapy. Cancer Res. 76:513–516, 2016.

    Google Scholar 

  47. Miranda-Goncalves, V., et al. Monocarboxylate transporters (MCTs) in gliomas: expression and exploitation as therapeutic targets. Neuro-oncology 15:172–188, 2013.

    Google Scholar 

  48. Morandi, F., et al. CD38: a target for immunotherapeutic approaches in multiple myeloma. Front. Immunol. 9:2722, 2018.

    Google Scholar 

  49. Moriarty, A. T., L. Wiersema, W. Snyder, P. K. Kotylo, and D. W. McCloskey. Immunophenotyping of cytologic specimens by flow cytometry. Diagn. Cytopathol. 9:252–258, 1993.

    Google Scholar 

  50. Nair, M. G., et al. Alternatively activated macrophage-derived RELM-α is a negative regulator of type 2 inflammation in the lung. J. Exp. Med. 206:937–952, 2009.

    Google Scholar 

  51. Nakao, S. et al. Intratumoral expression of IL-7 and IL-12 using an oncolytic virus increases systemic sensitivity to immune checkpoint blockade. Sci. Transl. Med. 12, https://doi.org/10.1126/scitranslmed.aax7992 (2020).

  52. Nasi, A., and B. Rethi. Disarmed by density A glycolytic break for immunostimulatory dendritic cells? Oncoimmunology 2013. https://doi.org/10.4161/onci.26744.

    Article  Google Scholar 

  53. Ngo, H., S. M. Tortorella, K. Ververis, and T. C. Karagiannis. The Warburg effect: molecular aspects and therapeutic possibilities. Mol. Biol. Rep. 42:825–834, 2015. https://doi.org/10.1007/s11033-014-3764-7.

    Article  Google Scholar 

  54. Nikolic, T., and B. Roep. Regulatory multitasking of tolerogenic dendritic cells–lessons taken from vitamin d3-treated tolerogenic dendritic cells. Front. Immunol. 4:113, 2013.

    Google Scholar 

  55. Nouri-Shirazi, M., et al. Dendritic cells capture killed tumor cells and present their antigens to elicit tumor-specific immune responses. J. Immunol. 165:3797–3803, 2000.

    Google Scholar 

  56. Ohashi, T., et al. M2-like macrophage polarization in high lactic acid-producing head and neck cancer. Cancer Sci. 108:1128–1134, 2017. https://doi.org/10.1111/cas.13244.

    Article  Google Scholar 

  57. Ohl, L., et al. CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity 21:279–288, 2004.

    Google Scholar 

  58. Ostuni, R., F. Kratochvill, P. J. Murray, and G. Natoli. Macrophages and cancer: from mechanisms to therapeutic implications. Trends Immunol. 36:229–239, 2015. https://doi.org/10.1016/j.it.2015.02.004.

    Article  Google Scholar 

  59. Ovens, M. J., A. J. Davies, M. C. Wilson, C. M. Murray, and A. P. Halestrap. AR-C155858 is a potent inhibitor of monocarboxylate transporters MCT1 and MCT2 that binds to an intracellular site involving transmembrane helices 7-10. Biochem. J. 425:523–530, 2010. https://doi.org/10.1042/bj20091515.

    Article  Google Scholar 

  60. Pearce, E. J., and B. Everts. Dendritic cell metabolism. Nat. Rev. Immunol. 15:18–29, 2015.

    Google Scholar 

  61. Randolph, G. J., V. Angeli, and M. A. Swartz. Dendritic-cell trafficking to lymph nodes through lymphatic vessels. Nat. Rev. Immunol. 5:617–628, 2005.

    Google Scholar 

  62. Ranganathan, P., et al. GPR81, a cell-surface receptor for lactate, regulates intestinal homeostasis and protects mice from experimental colitis. J. Immunol. 200:1781–1789, 2018.

    Google Scholar 

  63. Samuvel, D. J., K. P. Sundararaj, A. Nareika, M. F. Lopes-Virella, and Y. Huang. Lactate boosts TLR4 signaling and NF-κB pathway-mediated gene transcription in macrophages via monocarboxylate transporters and MD-2 up-regulation. J. Immunol. 182:2476–2484, 2009.

    Google Scholar 

  64. Shiraishi, D., et al. CD163 is required for protumoral activation of macrophages in human and murine sarcoma. Cancer Res. 78:3255–3266, 2018.

    Google Scholar 

  65. Singh-Jasuja, H., et al. The mouse dendritic cell marker CD11c is down-regulated upon cell activation through Toll-like receptor triggering. Immunobiology 218:28–39, 2013.

    Google Scholar 

  66. Sozzani, S., et al. Cutting edge: differential regulation of chemokine receptors during dendritic cell maturation: a model for their trafficking properties. J. Immunol. 161:1083–1086, 1998.

    Google Scholar 

  67. Stone, S. C., et al. Lactate secreted by cervical cancer cells modulates macrophage phenotype. J. Leukoc. Biol. 105:1041–1054, 2019.

    Google Scholar 

  68. Takashiba, S., et al. Differentiation of monocytes to macrophages primes cells for lipopolysaccharide stimulation via accumulation of cytoplasmic nuclear factor κB. Infect. Immun. 67:5573–5578, 1999.

    Google Scholar 

  69. Tan, Z., et al. The monocarboxylate transporter 4 is required for glycolytic reprogramming and inflammatory response in macrophages. J. Biol. Chem. 290:46–55, 2015. https://doi.org/10.1074/jbc.M114.603589.

    Article  Google Scholar 

  70. Tourkova, I. L., Z. R. Yurkovetsky, M. R. Shurin, and G. V. Shurin. Mechanisms of dendritic cell-induced T cell proliferation in the primary MLR assay. Immunol. Lett. 78:75–82, 2001.

    Google Scholar 

  71. Umansky, V. Immunosuppression in the tumor microenvironment: Where are we standing? Semin. Cancer Biol. 22:273–274, 2012. https://doi.org/10.1016/j.semcancer.2012.05.001.

    Article  Google Scholar 

  72. Veglia, F., and D. I. Gabrilovich. Dendritic cells in cancer: the role revisited. Curr. Opin. Immunol. 45:43–51, 2017. https://doi.org/10.1016/j.coi.2017.01.002.

    Article  Google Scholar 

  73. Vermeulen, M., et al. Acidosis improves uptake of antigens and MHC class I-restricted presentation by dendritic cells. J. Immunol. 172:3196–3204, 2004.

    Google Scholar 

  74. Villadangos, J. A., and P. Schnorrer. Intrinsic and cooperative antigen-presenting functions of dendritic-cell subsets in vivo. Nat. Rev. Immunol. 7:543–555, 2007.

    Google Scholar 

  75. Walenta, S. & Mueller-Klieser, (2017) W. F. in Semin. Radiat. Oncol. 267-274 (Elsevier).

  76. Walenta, S., T. Schroeder, and W. Mueller-Klieser. Lactate in solid malignant tumors: potential basis of a metabolic classification in clinical oncology. Curr. Med. Chem. 11:2195–2204, 2004.

    Google Scholar 

  77. Wang, Y.-C., et al. Lipopolysaccharide-induced maturation of bone marrow-derived dendritic cells is regulated by notch signaling through the up-regulation of CXCR4. J. Biol. Chem. 284:15993–16003, 2009.

    Google Scholar 

  78. Wang, Y., et al. High expression of CD11c indicates favorable prognosis in patients with gastric cancer. World J. Gastroenterol. W.J.G. 21:9403, 2015.

    Google Scholar 

  79. Worbs, T., et al. Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells. J. Exp. Med. 203:519–527, 2006.

    Google Scholar 

  80. Wu, A. A., V. Drake, H. S. Huang, S. C. Chiu, and L. Zheng. Reprogramming the tumor microenvironment: tumor-induced immunosuppressive factors paralyze T cells. Oncoimmunology 2015. https://doi.org/10.1080/2162402x.2015.1016700.

    Article  Google Scholar 

  81. Xuan, W., Q. Qu, B. Zheng, S. Xiong, and G. H. Fan. The chemotaxis of M1 and M2 macrophages is regulated by different chemokines. J. Leukoc. Biol. 97:61–69, 2015.

    Google Scholar 

  82. Xue, Q., Y. Yan, R. Zhang, and H. Xiong. Regulation of iNOS on immune cells and its role in diseases. Int. J. Mol. Sci. 19:3805, 2018.

    Google Scholar 

  83. Zaveri, T. D., J. S. Lewis, N. V. Dolgova, M. J. Clare-Salzler, and B. G. Keselowsky. Integrin-directed modulation of macrophage responses to biomaterials. Biomaterials 35:3504–3515, 2014.

    Google Scholar 

  84. Zorina, Z. A., and T. A. Obozova. New data on the brain and cognitive abilities of birds. Zool. Z. 90:784–802, 2011.

    Google Scholar 

Download references

ACKNOWLEDGMENTS

This work was supported by the National Institutes of Health (Grants R35125012 and R01AI139399).

Author Contributions

RS contributed to the design and execution of experiments, analysis of data, and compilation of the manuscript. BT and HH contributed to the execution of experiments and analysis of data. NP contributed to the analysis of RNA sequencing data and discussion on gene expression. RA provided input on experimental protocols. JSL contributed to the design and execution of experiments, analysis of data, manuscript compilation and has primary responsibility for the content of the manuscript.

Conflict of interest

Rapeepat Sangsuwan, Bhasirie Thuamsang, Noah J. Pacifici, Hyunsoo Han, Svetlana Miakicheva, Riley Allen, and Jamal S. Lewis have no conflicts of interest to disclose.

Research Involving Human and Animal Rights

C57BL/6 and BALB/cByJ mice were purchased from Jackson Laboratories and were housed in specific pathogen-free environment conditions at the University of California, Davis TRACS facility and used according to the UC Davis Institutional Animal Care and Use Committee (IACUC).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jamal S. Lewis.

Additional information

Associate Editor Shelly Peyton oversaw the review of this article.

Publisher's Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 2058 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sangsuwan, R., Thuamsang, B., Pacifici, N. et al. Lactate Exposure Promotes Immunosuppressive Phenotypes in Innate Immune Cells. Cel. Mol. Bioeng. 13, 541–557 (2020). https://doi.org/10.1007/s12195-020-00652-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12195-020-00652-x

Keywords

Navigation