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
. 2021 Oct 14;20(1):133.
doi: 10.1186/s12943-021-01420-9.

AMPK promotes antitumor immunity by downregulating PD-1 in regulatory T cells via the HMGCR/p38 signaling pathway

Affiliations

AMPK promotes antitumor immunity by downregulating PD-1 in regulatory T cells via the HMGCR/p38 signaling pathway

Ram Hari Pokhrel et al. Mol Cancer. .

Abstract

Background: AMP-activated protein kinase (AMPK) is a metabolic sensor that maintains energy homeostasis. AMPK functions as a tumor suppressor in different cancers; however, its role in regulating antitumor immunity, particularly the function of regulatory T cells (Tregs), is poorly defined.

Methods: AMPKα1fl/flFoxp3YFP-Cre, Foxp3YFP-Cre, Rag1-/-, and C57BL/6 J mice were used for our research. Flow cytometry and cell sorting, western blotting, immuno-precipitation, immuno-fluorescence, glycolysis assay, and qRT-PCR were used to investigate the role of AMPK in suppressing programmed cell death 1 (PD-1) expression and for mechanistic investigation.

Results: The deletion of the AMPKα1 subunit in Tregs accelerates tumor growth by increasing the expression of PD-1. Metabolically, loss of AMPK in Tregs promotes glycolysis and the expression of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), a key enzyme of the mevalonate pathway. Mechanistically, AMPK activates the p38 mitogen-activated protein kinase (MAPK) that phosphorylates glycogen synthase kinase-3β (GSK-3β), inhibiting the expression of PD-1 in Tregs.

Conclusion: Our study identified an AMPK regulatory mechanism of PD-1 expression via the HMGCR/p38 MAPK/GSK3β signaling pathway. We propose that the AMPK activator can display synergic antitumor effect in murine tumor models, supporting their potential clinical use when combined with anti-PD-1 antibody, anti-CTLA-4 antibody, or a HMGCR inhibitor.

Keywords: AICAR; AMPK; Compound C; HMGCR; PD-1; Tregs; Tumor.

PubMed Disclaimer

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
AMPK deficiency in Tregs promotes tumor growth. B16F10 melanoma cells were injected subcutaneously into C57BL/6 J mice and analyzed after 22 days. A Analysis of total LKB1 and AMPK in Tregs obtained from tumor-free and tumor-bearing C57BL/6 J mice by western blotting. B Detection of the mRNA expression of the prkaa1 gene by real-time PCR in Tregs isolated from tumor-free and tumor-bearing C57BL/6 J mice. The C tumor volume and D Tumor weight in WT and AMPKfl/flFoxp3-Cre mice injected s.c. with B16F10 melanoma cells. E Flow cytometric analysis of the percentage of CD4+ and CD8+ T cells; CD4+Foxp3+ Tregs and Tregs cellularity in the tumors of WT and AMPKfl/flFoxp3-Cre mice. F Analysis of IFN-γ-producing CD4+ and CD8+ T cells percentage in tumors from WT and AMPKfl/flFoxp3-Cre mice by flow cytometry. G Flow cytometry analysis of GZB-producing CD8+ T cells in tumors from WT and AMPKfl/flFoxp3-Cre mice. H Evaluation of in vivo cytolytic activity in the spleen and draining lymph node of WT and AMPKfl/flFoxp3-Cre mice. The data are presented as the mean ± standard deviation (SD); n = 5 mice per group. *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 2
Fig. 2
AMPK deficiency in Tregs upregulates the expression of PD-1. Bar diagram representation of flow cytometric analysis of relative mean fluorescence intensities (MFI) of PD-1, CTLA-4, Nrp1, ICOS, GITR, OX40, CD73, and CD39 in Tregs A gated from splenocytes of tumor-free mice and B gated from tumor-infiltrated lymphocytes. MFI of PD-1, Nrp1, and ICOS in splenic Tregs from WT mice after treatment with C compound C or D AICAR for 24 h. E Immunoblot analysis of PD-1 in Tregs treated with AICAR and compound C at the indicated doses and times of stimulation with anti-CD3/CD28. F T cell suppression assays with WT and AMPK-KO Tregs. G Bar diagram representation of flow cytometric analysis of IL-10-expressing WT and AMPK-KO Tregs and qRT-PCR results of the Il-10 mRNA levels. H MFI of TGF-β-expressing WT and AMPK- KO Tregs and qRT-PCR results of the Tgf-β mRNA levels. The data are presented as the mean ± standard deviation (SD); n = 5 mice per group. *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 3
Fig. 3
Treatment with anti-PD-1 antibody reduces tumor growth in AMPKfl/flFoxp3-Cre mice. B16F10 melanoma tumor cells were injected subcutaneously into AMPK-WT and AMPKfl/flFoxp3-Cre mice. Anti-mouse PD-1 was administered i.p. at 200 μg/mouse at 8, 11, 14, and 17 days after the injection of cancer cells. Tumors were analyzed at day 20 post-tumor transplantation. The A volume and B weight of the tumors from WT and AMPKfl/flFoxp3-Cre mice treated with or without anti-PD-1 antibody. C Percentage of CD4+ and CD8+ T cells and CD4+Foxp3+ Tregs in the tumors of WT and AMPKfl/flFoxp3-Cre mice treated with or without anti-PD-1 antibody. D Flow cytometry analysis of the percentage of GZB- and IFN-γ-producing CD8 + T cells in lymphocytes isolated from tumor tissues. E Bar diagram representation of flow cytometric analysis of the percentage of Foxp3+IL-10+ T cells. F Flow cytometric analysis and MFI of ICOS and Nrp1 in the tumors of WT and AMPKfl/flFoxp3-Cre mice with or without anti-PD-1 treatment. The data are presented as the mean ± standard deviation (SD); n = 5 mice per group. *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 4
Fig. 4
AMPK activation negatively regulates HMGCR in Treg cells. A Analysis of the indicated genes in Tregs isolated from WT and AMPKfl/flFoxp3-Cre mice by RT PCR. B Immunoblot analysis of the indicated proteins in WT and AMPK-KO Tregs after 1 h of resting and 4 h of stimulation with anti-CD3/CD28. C Detection of the phosphorylated and dephosphorylated form of HMGCR from WT Tregs treated with AICAR at indicated doses after 4 h of stimulation with anti-CD3/CD28. D Immunoprecipitation of the lysates of WT Tregs using anti-AMPK and anti-HMGCR antibodies. E The volume and weight of B16F10 tumors from WT mice after treatment with PBS, AICAR, statin, and AICAR + statin combination therapy. F The volume and weight of B16F10 tumors in Rag1−/− mice following PBS treatment or co-treatment with AICAR and statin. G MFI of PD-1 in Tregs from tumor-bearing WT mice after the indicated treatments. The data are presented as the mean ± standard deviation (SD); n = 5 mice per group. *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 5
Fig. 5
AMPK activates p38 MAPK through the inhibition of HMGCR to regulate the expression of PD-1. A Western blot analysis of the expression of PD-1 in treatment with the AMPK modulators: AICAR and compound C and the mevalonate pathway byproducts: cholesterol, GGPP, and mevalonate. B Immunoblot analysis of phosphorylated and total ERK, JNK, and p38 MAPK in Tregs from WT and AMPKfl/flFoxp3-Cre mice after 1 h of resting and 3 h of stimulation with anti-CD3/CD28. Western blot analysis of phosphorylated and total p38 MAPK in WT Tregs treated with or without C AICAR or D compound C at the indicated doses. E Immunoblot analysis of phosphorylated and total ERK and JNK in WT Tregs treated with AICAR, compound C, or statin. F Immunoblot analysis of phosphorylated and total form of p38 in Tregs treated with different concentrations of statin. G Immunoblot analysis of PD-1 in Tregs treated with SB203580 at the indicated doses. H MFI of PD-1 in WT Tregs with or without SB203580 treatment. The I tumor volume and J weight of B16F10 melanoma tumors in WT mice treated with PBS or SB203580. K MFI of PD-1 in Tregs isolated from tumor tissues after treatment with PBS or SB203580. The data are presented as the mean ± standard deviation (SD); n = 5 mice per group. *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 6
Fig. 6
AMPK suppresses the activity of GSK3β through p38 MAPK activation to regulate the expression of PD-1. A Analysis of phosphorylated and total GSK3β and β-catenin in WT and AMPK-KO Tregs stimulated with or without anti-CD3/CD28 stimulation for 4 h. Analysis of the expression of GSK3β and β-catenin by western blotting in activated Tregs after B AICAR and C compound C treatment at the indicated doses. D Immunoblot analysis of the indicated proteins in activated Tregs treated with the p38 MAPK inhibitor SB203580 at the indicated doses. E Immunoprecipitation assay of the lysates of WT Tregs using anti-p38 and anti-GSK3β antibodies. Flow cytometric analysis of F PD-1 and G T-bet in Tregs from WT and AMPKfl/flFoxp3-Cre mice after i.p. treatment with or without the GSK3β inhibitor SB216763 daily for 7 days (2 μg/kg). The data are presented as the mean ± standard deviation (SD); n = 5 mice per group. *P < 0.05; **P < 0.01; ***P < 0.001

References

    1. DeBerardinis RJ, Chandel NS. Fundamentals of cancer metabolism. Sci Adv. 2016;2:e1600200. doi: 10.1126/sciadv.1600200. - DOI - PMC - PubMed
    1. Buck MD, Sowell RT, Kaech SM, Pearce EL. Metabolic instruction of immunity. Cell. 2017;169:570–586. doi: 10.1016/j.cell.2017.04.004. - DOI - PMC - PubMed
    1. Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017;27:109–118. doi: 10.1038/cr.2016.151. - DOI - PMC - PubMed
    1. Wang Y-a, Li X-L, Mo Y-Z, Fan C-M, Tang L, Xiong F, Guo C, Xiang B, Zhou M, Ma J. Effects of tumor metabolic microenvironment on regulatory T cells. Mol Cancer. 2018;17:168. doi: 10.1186/s12943-018-0913-y. - DOI - PMC - PubMed
    1. Sierra RA, Thevenot P, Raber PL, Cui Y, Parsons C, Ochoa AC, Trillo-Tinoco J, Del Valle L, Rodriguez PC. Rescue of notch-1 signaling in antigen-specific CD8+ T cells overcomes tumor-induced T-cell suppression and enhances immunotherapy in cancer. Cancer Immunol Res. 2014;2:800–811. doi: 10.1158/2326-6066.CIR-14-0021. - DOI - PMC - PubMed

Publication types

MeSH terms

Substances

LinkOut - more resources