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
. 2023 Oct 27;14(1):6858.
doi: 10.1038/s41467-023-42634-3.

Mitochondrial dysfunction promotes the transition of precursor to terminally exhausted T cells through HIF-1α-mediated glycolytic reprogramming

Affiliations

Mitochondrial dysfunction promotes the transition of precursor to terminally exhausted T cells through HIF-1α-mediated glycolytic reprogramming

Hao Wu et al. Nat Commun. .

Abstract

T cell exhaustion is a hallmark of cancer and persistent infections, marked by inhibitory receptor upregulation, diminished cytokine secretion, and impaired cytolytic activity. Terminally exhausted T cells are steadily replenished by a precursor population (Tpex), but the metabolic principles governing Tpex maintenance and the regulatory circuits that control their exhaustion remain incompletely understood. Using a combination of gene-deficient mice, single-cell transcriptomics, and metabolomic analyses, we show that mitochondrial insufficiency is a cell-intrinsic trigger that initiates the functional exhaustion of T cells. At the molecular level, we find that mitochondrial dysfunction causes redox stress, which inhibits the proteasomal degradation of hypoxia-inducible factor 1α (HIF-1α) and promotes the transcriptional and metabolic reprogramming of Tpex cells into terminally exhausted T cells. Our findings also bear clinical significance, as metabolic engineering of chimeric antigen receptor (CAR) T cells is a promising strategy to enhance the stemness and functionality of Tpex cells for cancer immunotherapy.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1. T cell exhaustion is characterized by metabolic reprogramming.
a C57BL/6 mice were chronically infected with the LCMV strain clone 13 (LCMVCL13) and CD8+CD44+PD-1hi T cells were subjected to single-cell RNA sequencing 21 days post infection. Uniform manifold approximation and projection (UMAP) visualization of ~ 13.000 non-proliferating (Mki67 negative) T cells colored based on their classification into six clusters. b Prediction of developmental trajectories using slingshot analysis; cells are color-coded according to pseudotime. c Normalized gene expression of Tcf7, Slamf6 (Ly108), Sell (CD62L), Havcr2 (Tim-3), Cxcr6 and Gzma projected onto UMAP clusters. d Dot plot analysis of selected markers representing Tpex and Tex cell subsets; color intensity and dot size represent z-score mean expression and percentage of cells expressing the gene, respectively. e Enrichment of mitochondrial and respiratory chain gene expression signatures in clusters representing Tpex, CTL-like and terminally exhausted (Tex) cells. f Flow cytometric cell sorting strategy of Tpex and Tex cells from chronically infected mice. g Analyzes of oxygen consumption rate (OCR) and spare respiratory capacity (SRC) of Tpex (n = 9 mice) and Tex cells (n = 4 mice) using a Seahorse extracellular flux analyzer; means ± SEM. h Analysis of mitochondrial content in Tpex and Tex cells using LCMVCL13 infected mito-Dendra2 mice; means ± SEM of 3 mice. i Mitochondrial regeneration of mito-Dendra2 Tpex and Tex cells after photoconversion in vitro; means ± SEM of 7 mice. j Glycolytic proton efflux rate (glycoPER) analyzes of Tpex (n = 5 mice) and Tex cells (n = 2 mice) using a Seahorse extracellular flux analyzer; means ± SEM. k Violin plots displaying Slc2a3 (GLUT3), Aldoa and Gapdh gene expression in Tpex and Tex cell clusters as shown in (a). l Ratio of extracellular acidification rate (ECAR) to OCR of FACS-sorted Ly108+ Tpex and Tim3+ Tex isolated from chronically infected mice ex vivo; means ± SEM of 4-8 mice. m Relative contribution of glycolysis and mitochondrial respiration to cellular ATP production in naïve (n = 10 mice), Tpex (n = 6 mice) and Tex cells (n = 4 mice). Unpaired two-tailed Student’s t-test in (g-i), (k), (l) and two-way ANOVA in (e).
Fig. 2
Fig. 2. Genetic suppression of mitochondrial ATP production promotes glycolytic-transcriptional reprogramming of T cells.
a, b Analyzes of (a) oxygen consumption rate (OCR) and (b) glycolytic proton efflux rate (glycoPER) of WT and mPiC-deficient (Slc25a3fl/flCd4Cre) T cells at day 2 of culture using a Seahorse extracellular flux analyzer; means ± SEM of 7 mice. c Analysis of ATP concentrations in unstimulated and anti-CD3/CD28 activated WT and mPiC-deficient T cells; means ± SEM of 6 mice. d Relative contribution of glycolysis and mitochondrial respiration to cellular ATP production in anti-CD3/CD28 stimulated WT and mPiC-deficient T cells; means of 3 mice. e Network clustering of RNA sequencing data of significantly (p < 0.05) enriched gene expression signatures between CTL-differentiated WT and mPiC-deficient T cells. Down- and upregulated gene signatures in mPiC-deficient T cells are shown in blue and red, respectively. f, g Gene set enrichment analysis (GSEA) of (f) oxidative phosphorylation (KEGG pathway) and (g) effector versus exhausted T cells (GSE9650) gene signatures in differentiated WT and mPiC-deficient T cells after 6 days of culture. h Heatmap expression analysis of selected genes in WT and mPiC-deficient CTLs after 6 days in culture. ik Differentiation of WT and mPiC-deficient T cells in vitro, means ± SEM of 5 mice. j, k Flow cytometric quantification of (j) exhaustion and (k) memory marker expression in WT and mPiC-deficient CTLs, means ± SEM of 5 mice. l Analysis of polyfunctional TNFα, IFNγ and IL-2 expression by WT and mPiC-deficient T cells after 6 days of culture and anti-CD3/CD28 restimulation; means ± SEM of 3 mice. m Analysis of apoptosis in resting and anti-CD3/CD28 stimulated T cells by flow cytometry; means ± SEM of 3 mice. Two-tailed unpaired Student’s t-test in (ac) and (im).
Fig. 3
Fig. 3. Mitochondrial respiration controls the functional exhaustion of virus-specific T cells.
a Adoptive co-transfer of GFP+ WT and tdTomato+ mPiC-deficient (Slc25a3fl/flCd4Cre) P14 T cells into C57BL/6 mice before chronic infection with LCMV clone 13 (LCMVCL13). Flow cytometric analysis of Tpex and Tex cells in spleen and LNs of the host mice 14 days post infection (d.p.i.); n = 8 mice. b Acute infection of WT and Slc25a3fl/flCd4Cre mice with LCMV Armstrong (LCMVARM). Analysis of Tpex and Tex cells was performed 10 d.p.i.; means ± SEM of 5 mice. c, d Analysis of TNFα, IFNγ, and IL-2 expression after PMA/iono restimulation of WT and mPiC-deficient P14 T cells after co-transfer into chronically infected mice; means ± SEM of 6–10 mice. em Ectopic expression of mPiC attenuates T cell exhaustion. e Retroviral transduction of WT P14 T cells with GFP+ SLC25A3/mPiC or Ametrine+ empty vector followed by adoptive co-transfer into chronically infected mice. f Representative flow cytometric analysis of GFP+ and Ametrine+ P14 T cells after transfer into LCMVCL13 infected CD45.1+ mice. g, h Relative and absolute numbers of GFP+ and Ametrine+ P14 T cells in the spleens; means ± SEM of 2-6 mice. i, j Basal and maximal oxygen consumption rate (OCR) (i) and mitochondrial ATP production rate (j) in mPiC (GFP+) and empty vector (Ametrine+) transduced P14 cells ex vivo 7 d.p.i. using a Seahorse extracellular flux analyzer; means ± SEM of 3 mice analyzed in 2-3 technical replicates. k Ratio of OCR to extracellular acidification rate (ECAR) in mPiC overexpressing versus empty vector T cells; means ± SEM of 3 mice analyzed in 5 technical replicates. l Flow cytometric analysis of Tpex and Tex cell ratio in mPiC (GFP+) and empty vector transduced (Ametrine+) P14 cells 7 d.p.i.; n = 6 mice. m IFNγ and IL-2 expression in mPiC and empty vector transduced P14 cells after restimulation 7 d.p.i.; means ± SEM of 6 mice. Paired and two-tailed unpaired Student’s t-test in (a), (b), (d), and (im) or 2-way ANOVA in (g, h).
Fig. 4
Fig. 4. Mitochondrial insufficiency causes ROS-mediated HIF-1 α protein stabilization.
a Volcano plot of differential metabolite concentrations of in vitro activated WT and mPiC-deficient (Slc25a3fl/flCd4Cre) T cells analyzed by untargeted liquid chromatography and mass spectrometry (LC/MS); means ± SEM of 4 mice. b Analysis of NADPH/NADP+ ratios in WT and mPiC-deficient T cells using LC/MS; means ± SEM of 4 mice. c Gene set enrichment analysis (GSEA) of the reactive oxygen species (ROS) pathway gene signature in WT and mPiC-deficient T cells. d Heatmap analysis of selected genes involved in ROS detoxification. e Flow cytometric analysis of cellular (CellROX) and mitochondrial ROS (MitoSox) in WT and mPiC-deficient T cells; means ± SEM of 5 mice. f Scavenging of ROS by N-acetylcysteine (NAC) prevents exhaustion of mPiC-deficient T cells. Flow cytometric analyzes of CellRox, PD-1, Tim-3 and granzyme A expression in WT and mPiC-deficient T cells treated with NAC; means ± SEM of 4–8 mice. g Analysis of TNFα, IFNγ and IL-2 expression after PMA/iono restimulation of mPiC-deficient T cells treated with or without NAC; means ± SEM of 5 mice. h Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis using bulk RNA sequencing data of WT and mPiC-deficient T cells. i Upstream transcription factor prediction analysis using differentially expressed genes (DEGs) between WT and mPiC-deficient T cells. j Analysis of Hif1a gene expression in WT and mPiC-deficient T cells by RNA sequencing; means ± SEM of 3 mice. k Flow cytometric analysis of HIF-1α protein expression in WT, mPiC- and HIF-1α-deficient T cells; means ± SEM of 3 mice. l, m Differentiation of WT, mPiC-deficient (Slc25a3fl/flCd4Cre) and mPiC/HIF-1α double-deficient (Slc25a3fl/flHif1afl/flCd4Cre) T cells in vitro. Representative flow cytometric analysis (l) and quantification of PD-1, Tim-3, granzyme A, Lag3 and Ly108 expression on WT, mPiC-deficient and mPiC/HIF-1α double-deficient T cells (m); means ± SEM of 2–7 mice. Two-tailed unpaired Student’s t-test in (b), (e–g), (k) and (m).
Fig. 5
Fig. 5. HIF-1α controls terminal differentiation of virus-specific T cells.
a Adoptive co-transfer of WT (tdTomato+GFP+) and HIF-1α-deficient (tdTomato+) P14 T cells into C57BL/6 mice before chronic infection with LCMV clone 13 (LCMVCL13). 21 days post infection (d.p.i.), donor P14 WT and HIF-1α-deficient T cells were FACS sorted, barcoded, and multiplexed in a 1:1 ratio and subjected to single-cell RNA sequencing. Uniform manifold approximation and projection (UMAP) visualization of ~8.400 non-proliferating (Mki67 low/negative) T cells identified four clusters. Prediction of developmental trajectories using slingshot analysis; color-coding according to pseudotime. b Normalized gene expression of Tcf7, Slamf6 (Ly108), Sell (CD62L), Havcr2 (Tim-3), Cxcr6 and Gzma projected onto UMAP clusters. c Dot plot analysis of selected cluster markers representing Tpex and Tex cell subsets; color intensity and dot size represent z-score mean expression and percentage of cells expressing the gene, respectively. d Relative contribution of individual UMAP clusters in WT and Hif-1α-deficient P14 T cells. e Volcano plot of differentially expressed genes (DEGs) between WT and HIF-1α-deficient P14 T cells using scRNA sequencing. f Adoptive co-transfer of WT (GFP+) and HIF-1α-deficient (tdTomato+) P14 T cells into C57BL/6 mice before chronic infection with LCMVCL13. Flow cytometric analysis of Tpex and Tex cells was performed 14 d.p.i.; n = 12 mice. Two-tailed paired Student’s t-test in (f).
Fig. 6
Fig. 6. HIF-1α-mediated glycolytic reprogramming promotes T cell exhaustion.
a KEGG pathway enrichment analysis using single-cell RNA sequencing of WT and HIF-1α-deficient T cells. b, c Violin plots displaying Pfkl, Aldoa, Tpi1, Gapdh, Eno1, and Pkm gene expression in Tpex (b) and Tex cells (c) of WT and HIF-1α-deficient (Hif1afl/flCd4Cre) P14 T cells analyzed by single-cell RNA sequencing. d Differentiation of WT and HIF-1α-deficient T cells under hypoxia in vitro, means ± SEM of 6 mice. e, f Analyzes of glycolytic proton efflux rate (glycoPER) (e) and oxygen consumption rate (OCR) (f) in WT and HIF-1α-deficient T cells using a Seahorse extracellular flux analyzer; means ± SEM of 3 mice. g Relative contribution of glycolysis and mitochondrial respiration to cellular ATP production in WT and HIF-1α-deficient T cells; means ± SEM of 3 mice. h, i Inhibition of glycolysis sustains the stemness of virus-specific T cells. h Adoptive co-transfer of 2-DG treated (tdTomato+ GFP+) and control (GFP+) P14 T cells into C57BL/6 mice after chronic infection with LCMVCL13. Flow cytometric analysis of Tpex and Tex cells among donor P14 cells 10 d.p.i.; n = 6 mice. i Analysis of TNFα, IFNγ, and IL-2 expression by 2-DG-treated and control P14 T cells 10 days after co-transfer into chronically infected mice; means ± SEM of 6 mice. jl 2-DG treatment augments the antitumor efficacy of CAR T cells. j A total of 1 × 106 anti-hCD19 CAR T cells treated with or without 2-DG for 24 h were adoptively transferred into Rag1–/– mice 7 days after MC38 tumor inoculation. k, l Analysis of tumor growth (k) and cumulative survival (l) of tumor-bearing host mice after transfer of 2-DG or control-treated CAR T cells; means ± SEM of 9–11 mice. Unpaired and two-tailed paired Student’s t-test in (bd) and (h). In (k) and (l), 2-way ANOVA and Mantel-Cox test, respectively.

Comment in

References

    1. McLane LM, Abdel-Hakeem MS, Wherry EJ. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 2019;37:457–495. doi: 10.1146/annurev-immunol-041015-055318. - DOI - PubMed
    1. Franco F, Jaccard A, Romero P, Yu YR, Ho PC. Metabolic and epigenetic regulation of T-cell exhaustion. Nat. Metab. 2020;2:1001–1012. doi: 10.1038/s42255-020-00280-9. - DOI - PubMed
    1. Zehn D, Thimme R, Lugli E, de Almeida GP, Oxenius A. ‘Stem-like’ precursors are the fount to sustain persistent CD8(+) T cell responses. Nat. Immunol. 2022;23:836–847. doi: 10.1038/s41590-022-01219-w. - DOI - PubMed
    1. Utzschneider DT, et al. Early precursor T cells establish and propagate T cell exhaustion in chronic infection. Nat. Immunol. 2020;21:1256–1266. doi: 10.1038/s41590-020-0760-z. - DOI - PubMed
    1. Im SJ, et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature. 2016;537:417–421. doi: 10.1038/nature19330. - DOI - PMC - PubMed

Publication types

Substances