Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Sep 6;13(1):4981.
doi: 10.1038/s41467-022-32521-8.

Lactate increases stemness of CD8 + T cells to augment anti-tumor immunity

Affiliations

Lactate increases stemness of CD8 + T cells to augment anti-tumor immunity

Qiang Feng et al. Nat Commun. .

Abstract

Lactate is a key metabolite produced from glycolytic metabolism of glucose molecules, yet it also serves as a primary carbon fuel source for many cell types. In the tumor-immune microenvironment, effect of lactate on cancer and immune cells can be highly complex and hard to decipher, which is further confounded by acidic protons, a co-product of glycolysis. Here we show that lactate is able to increase stemness of CD8+ T cells and augments anti-tumor immunity. Subcutaneous administration of sodium lactate but not glucose to mice bearing transplanted MC38 tumors results in CD8+ T cell-dependent tumor growth inhibition. Single cell transcriptomics analysis reveals increased proportion of stem-like TCF-1-expressing CD8+ T cells among intra-tumoral CD3+ cells, a phenotype validated by in vitro lactate treatment of T cells. Mechanistically, lactate inhibits histone deacetylase activity, which results in increased acetylation at H3K27 of the Tcf7 super enhancer locus, leading to increased Tcf7 gene expression. CD8+ T cells in vitro pre-treated with lactate efficiently inhibit tumor growth upon adoptive transfer to tumor-bearing mice. Our results provide evidence for an intrinsic role of lactate in anti-tumor immunity independent of the pH-dependent effect of lactic acid, and might advance cancer immune therapy.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1. Lactate augments antitumor immunity through CD8+ T cells.
a Treatment regimen for glucose (Glc) or lactate (Lac). Glucose (5 g/kg) or lactate (1.68 g/kg) was subcutaneously administrated daily from Day 8 after tumor inoculation. b Tumor growth curve of MC38 tumor model treated with glucose or lactate. C57BL/6 mice (n = 6) were inoculated with 1 × 106 MC38 tumor cells and treated with glucose or lactate. c, Tumor growth curves of MC38 tumor in B6.129S7-Rag1tm1Mom (Rag1−/−) mice treated with lactate. Rag1−/− mice (n = 7) were inoculated with 1 × 106 MC38 tumor cells and treated with lactate. CD8+ T cell (d), CD4+ T cell (e) and macrophage (f) depletion assay in MC38 tumor model. C57BL/6 mice (n = 6) were inoculated with 1 × 106 MC38 tumor cells and treated with lactate. Anti-CD8 (10 mg/kg) was administered on day 6 and then every three days until the end of the experiment. Anti-CD4 (10 mg/kg) or anti-CSF1R (20 mg/kg) was administered on day 3 and then every three days until the end of the experiment. Data are shown as means ± SEM. P value was determined by one-tail two-way ANOVA with correction using Geisser-Greenhouse method. Source data are provided in Source Data file.
Fig. 2
Fig. 2. Lactate but not glucose promotes antitumor immunity in multiple tumor models.
a Treatment regimen for immunotherapy in combination with glucose (Glc) or lactate (Lac). Glucose (5 g/kg) or lactate (1.68 g/kg) was administrated subcutaneously daily one day after the first dose of anti-PD-1 (aPD1, i.p. injection) or PC7A vaccine (Vax, s.c. injection) treatment. b Tumor growth and survival data of anti-PD-1 combined with glucose or lactate in MC38 tumor model. C57BL/6 mice (n = 6) were inoculated with 1 × 106 MC38 tumor cells and treated with anti-PD-1 (10 mg/kg, day 7 and 10) in combination with glucose or lactate. TF: tumor free. c Tumor growth curves of anti-PD-1 or PC7A vaccine combined with lactate or glucose in TC-1 tumor model. C57BL/6 mice (n = 6) were inoculated with 1.5 × 105 TC-1 tumor cells and treated with anti-PD-1 (10 mg/kg, day 11 and 14) or PC7A vaccine (0.5 μg E7 peptide, day 11, 16) in combination with glucose or lactate. d, Tumor growth curve of anti-PD-1 combined with lactate or glucose in B16F10 tumor model. C57BL/6 mice (n = 6) were inoculated with 1.5 × 105 B16F10 tumor cells and treated with anti-PD-1 (10 mg/kg, day 5 and 8) in combination with glucose or lactate. e CD8+ T cell depletion assay in MC38 tumor model. C57BL/6 mice (n = 5) were inoculated with 1 × 106 MC38 tumor cells and treated with anti-PD-1 (10 mg/kg, day 7 and 10) in combination with glucose or lactate. Anti-CD8 (10 mg/kg) was administered at day 6 and then every three days until the end of the experiment. Data are shown as means ± SEM. P-value was determined by logrank test (b) or one-tail two-way ANOVA with correction using Geisser-Greenhouse method (a, b–e). ns: not significant. Source data and summary of all P values are provided in Source Data file.
Fig. 3
Fig. 3. Lactate treatment increases infiltrating CD8+ T cells in MC38 tumors.
a Experimental design of single cell transcriptomic analysis of anti-PD-1 with or without lactate. C57BL/6 mice were inoculated with 1 × 106 MC38 tumor cells and treated with anti-PD-1 (10 mg/kg, day 14 and 17) and lactate (1.68 g/kg, s.c. daily from day 15 to 19). Tumor and tumor draining lymph nodes were harvested on day 20 and analyzed by single cell RNA sequencing using the 10x platform. b tSNE plot of T cell clusters with location and cell type information analyzed with Seurat v3.0.1. DLN: tumor draining lymph nodes. c Distribution of T cells from different treatments and expression of marker genes. d Significantly upregulated pathways in tumor infiltrating CD8+ T cells after lactate treatment by unbiased gene set enrichment analysis (gene set database: c2.cp.kegg.v7.2.symbols). e Validation of increased tumor infiltrating CD8+ T cells by flow cytometry. C57BL/6 mice (n = 5) were inoculated with 1 × 106 MC38 tumor cells and treated with anti-PD-1 (10 mg/kg, day 14 and 17) and lactate (1.68 g/kg, s.c. daily from day 15 to 19). MC38 tumors were harvested on day 20 and analyzed by flow cytometry. f Analysis of apoptosis markers of CD8+ T cells in tumor microenvironment by flow cytometry. Data are shown as means ± SEM. P value was determined by two-tail unpaired t-test (e, f). Source data are provided in Source Data file.
Fig. 4
Fig. 4. Lactate treatment increases stem-like CD8+ T cell population in MC38 tumors.
a Pseudotime trajectory of CD8+ T cells in tumors identifies the differentiation process and distinct states of CD8+ T cell subtypes. b Labeling of top marker genes on pseudotime trajectory identifies the cells in state 1 are stem-like T cells while those in state 7 are exhausted T cells. c Change of CD8+ T cell fraction with aPD1+Lac verses aPD1 treatment. RO/E Lac analysis showed lactate treatment increased the T cell populations in states 1 and 7. d Validation of increase in TCF1+ PD1+ CD8+ T cell population after lactate treatment by flow cytometry. C57BL/6 mice (n = 8) were inoculated with 1 × 106 MC38 tumor cells and treated with anti-PD-1 (10 mg/kg, day 14 and 17) and lactate (1.68 g/kg, s.c. daily from day 15 to 19). Tumors were harvested on day 20 and analyzed by flow cytometry. e, f Flow cytometry plot and quantification of TCF-1 expressions in tumor-infiltrating CD8+ T cells after different treatments. Samples are the same as described in D. Data are shown as means ± SEM. P value was determined by one-tail Chi-square test (c) or two-tail unpaired t-test (d, f). Source data are provided in Source Data file.
Fig. 5
Fig. 5. Lactate increases TCF-1 expression and reduces apoptosis of CD8+ T cells during ex vivo expansion.
a Experimental design of ex vivo OT-I CD8+ T cell expansion. Fresh splenocytes from C57BL/6-Tg(TcraTcrb)1100Mjb/J mice were primed with SIINFEKL peptide (1 μg/mL) and hIL-2 (50 U/mL) for two days and stimulated with anti-CD3 and anti-CD28 (0.5 μg/mL each) from day 3 to 8 with hIL-2 (30 U/mL). b, c Flow cytometry plots and quantification of TCF-1hiCXCR3hi population of OT-I CD8+ T cells on day 8 of ex vivo expansion (n = 4 biologically independent samples). d, e Quantification of percentage of apoptotic OT-I CD8+ T cells by flow cytometry on day 8 of ex vivo expansion (n = 4 biologically independent samples). f, Relative gene expression in OT-I CD8+ T cells on day 4 detected by RT-PCR (n = 3 biologically independent samples). g Experimental design of ex vivo expansion of CD8+ T cells from human PBMCs. PBMCs from cord blood were activated and cultured in the presence of anti-CD3 and anti-CD28 beads (T cell: Beads = 1: 1) supplemented with hIL-2 (30 U/mL). h, i Flow cytometry plots and quantification of TCF-1hiCXCR3hi population of human CD8+ T cells on day 8 of ex vivo expansion (n = 4 biologically independent samples). Data are shown as means ± SEM. P-value was determined by two-tail unpaired t-test (c, e, i) or one-tail two-way ANOVA without correction (f). Source data are provided in Source Data file.
Fig. 6
Fig. 6. Lactate increases TCF-1 expression through inhibition of histone deacetylases (HDAC).
a Potential mechanisms for lactate induced TCF-1 upregulation. b Heatmap of significantly changed metabolites in CD8+ T cells treated with or without 40 mM lactate. c Source of upregulated metabolites with 40 mM lactate treatment. d TCF-1 protein expression in CD8+ T cells treated by different metabolites, GPCR agonist or HDAC inhibitors (n = 3 biologically independent samples). e Gene expression of Tcf7 in CD8+ T cells treated by different metabolites, GPCR agonist or HDAC inhibitors. The concentration of different metabolites was the same as the highest concentration in d (n = 6 biologically independent samples). f Western blot of histone H3K27ac of CD8+ T cells cultured with or without 40 mM lactate (One representative data was shown from 3 independently repeated experiments). g Quantification of H3K27ac enrichment by CUT&RUN PCR at the Tcf7 super enhancer locus (n = 3 biologically independent samples). Data are shown as means ± SEM. P-value was determined by two-tail unpaired t-test without adjustment (d, e) or two-tail ratio paired t-test (g). Source data are provided in Source Data file.
Fig. 7
Fig. 7. CD8+ T cells expanded under high lactate condition show potent tumor growth inhibition in vivo.
a Treatment regimen of T cell receptor engineered T cell therapy (TCR-T). OT-I splenocytes were cultured ex vivo for 4 days with or without 40 mM sodium lactate in the culture medium. After purification with negative selection magnetic beads, CD8+ T cells were transferred to MC38-OVA tumor bearing mice. Average tumor size is above 100 mm3 at the time of cell transfer. b The growth curves of MC38-OVA tumor model were significantly inhibited after transfer of TCR-T (5 × 105 or 2 × 106) pretreated with sodium lactate (n = 5). c Analysis of tumor-infiltrating T cells after adoptive TCR-T cell transfer. Seven days after the cell transfer, tumor-infiltrating lymphocytes were analyzed by flow cytometry. df Flow cytometry plot and quantification of transferred (CD45.2+) and endogenous (CD45.1+) OVA-tetramer+ CD8+ T cells show lactate pretreatment significantly increased the number of transferred CD8+ T cells (CD45.2+) but not endogenous CD8+ T cells (CD45.1+) in the MC38-OVA tumors (n = 5). Data are shown as means ± SEM. P-value was determined by one-tail two-way ANOVA with correction using Tukey method (b, c) or one-tail one-way ANOVA with correction using Geisser-Greenhouse method (e, f). Source data are provided in Source Data file.

References

    1. Pearce EL, Pearce EJ. Metabolic pathways in immune cell activation and quiescence. Immunity. 2013;38:633–643. doi: 10.1016/j.immuni.2013.04.005. - DOI - PMC - PubMed
    1. Jung J, Zeng H, Horng T. Metabolism as a guiding force for immunity. Nat. Cell Biol. 2019;21:85–93. doi: 10.1038/s41556-018-0217-x. - DOI - PubMed
    1. O’Neill LAJ, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 2016;16:553–565. doi: 10.1038/nri.2016.70. - DOI - PMC - PubMed
    1. Chang CH, et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell. 2013;153:1239–1251. doi: 10.1016/j.cell.2013.05.016. - DOI - PMC - PubMed
    1. Wang C, et al. CD5L/AIM regulates lipid biosynthesis and restrains Th17 cell pathogenicity. Cell. 2015;163:1413–1427. doi: 10.1016/j.cell.2015.10.068. - DOI - PMC - PubMed

Publication types