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 Aug 4;14(1):4677.
doi: 10.1038/s41467-023-39571-6.

Targeting of SLC25A22 boosts the immunotherapeutic response in KRAS-mutant colorectal cancer

Affiliations

Targeting of SLC25A22 boosts the immunotherapeutic response in KRAS-mutant colorectal cancer

Qiming Zhou et al. Nat Commun. .

Abstract

KRAS is an important tumor intrinsic factor driving immune suppression in colorectal cancer (CRC). In this study, we demonstrate that SLC25A22 underlies mutant KRAS-induced immune suppression in CRC. In immunocompetent male mice and humanized male mice models, SLC25A22 knockout inhibits KRAS-mutant CRC tumor growth with reduced myeloid derived suppressor cells (MDSC) but increased CD8+ T-cells, implying the reversion of mutant KRAS-driven immunosuppression. Mechanistically, we find that SLC25A22 plays a central role in promoting asparagine, which binds and activates SRC phosphorylation. Asparagine-mediated SRC promotes ERK/ETS2 signaling, which drives CXCL1 transcription. Secreted CXCL1 functions as a chemoattractant for MDSC via CXCR2, leading to an immunosuppressive microenvironment. Targeting SLC25A22 or asparagine impairs KRAS-induced MDSC infiltration in CRC. Finally, we demonstrate that the targeting of SLC25A22 in combination with anti-PD1 therapy synergizes to inhibit MDSC and activate CD8+ T cells to suppress KRAS-mutant CRC growth in vivo. We thus identify a metabolic pathway that drives immunosuppression in KRAS-mutant CRC.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1. SLC25A22 knockout affects immune cell infiltration in KRAS-mutant CRC.
a Breeding of ApcMin/+ and lsl-KrasG12D/+Villin-Cre to give ApcMin/+KrasG12D/+ Villin-Cre mice. At 7 weeks, colon tumors were harvested for RNA-seq. b Immune-related signaling are enriched in colon tumors (n = 2) from ApcMin/+KrasG12D/+Villin-Cre compared to ApcMin/+ mice (n = 2). Rich factor is defined as the ratio of differentially expressed gene number to total gene number in a particular pathway. P value, Fisher’s exact test. c Flow cytometry of immune cell populations in ApcMin/+KrasG12D/+Villin-Cre (n = 4) and ApcMin/+ mice (n = 6). MDSC, including PMN-MDSC, were increased in ApcMin/+KrasG12D/+Villin-Cre mice, together with decreased total T cells and CD8+ T cells in tumors. Each dot represents an independent mouse. d Colon tumor organoids derived from ApcMin/+KrasG12D/+ or ApcMin/+KrasG12D/+Slc25a22−/− mice with intestinal Slc25a22 knockout were implanted into C57BL/6 mice. Tumor growth was inhibited in Slc25a22 knockout organoids (n = 4). Each dot represents an independent mouse. e Slc25a22 knockout inhibited MDSC and PMN-MDSC, but induced total and CD8+ T cells (n = 4). Each dot represents an independent mouse. f SLC25A22 knockout (Slc-KO) in KRAS-mutant CT26 cells inhibited tumor growth in BALB/c mice (sgControl and SLC-KO1: n = 10 per group; SLC-KO2: n = 8). Each dot represents an independent tumor. g SLC25A22 knockout inhibited MDSC and PMN-MDSC, but increased total T cells and CD8+ T cells (n = 8). h Immunofluorescence and immunohistochemistry of immune cells in the tumors from ApcMin/+KrasG12D/+ and ApcMin/+KrasG12D/+Slc25a22−/− mice (n = 6), confirming reduced MDSC but increased CD8+ T cells by SLC25A22 knockout. Each dot represented the value from an independent capture field. Data are shown as mean ± SD (ch) and ± SEM for the growth curves (d, f). Two-tailed Student’s t test (ce, h). Two-tailed one-way ANOVA (f, g). Two-tailed two-way ANOVA for growth curve (d, f). ns, no significance. Source data are provided as a Source Data file.
Fig. 2
Fig. 2. SLC25A22 correlates with immune suppression in PBMC humanized mice and human KRAS-mutant CRC patients.
a The construction and validation of PBMC humanized mice model. b SLC25A22 knockout significantly inhibited the growth of DLD1 xenografts in PBMC humanized mice (sgControl: n = 15; SLC-KO1: n = 10). Each dot represents an independent tumor. c SLC25A22 knockout inhibited tumor infiltration of human MDSC (HLA-DR-CD11b+CD33+) while increasing T-cell (hCD3+CD4+/ hCD3+CD8+) infiltration (sgControl: n = 15; SLC-KO1: n = 10). Each dot represents an independent tumor. d Co-immunofluorescence of SLC25A22 and CD33 in a tissue microarray CRC cohort (n = 207). Positive correlation was revealed in only KRAS-mutant CRC (upper). Increased MDSC infiltration was found in SLC25A22-high CRC with mutant KRAS (lower). Each dot represents an independent patient. e In TCGA cohort (n = 383), SLC25A22 mRNA expression is significantly higher in MDSC-high tumors compared to MDSC-low tumors in KRAS-mutant CRC. Each dot represents an independent patient. f Immunohistochemistry of CD8 and SLC25A22 in CRC tissue microarray (n = 202). CD8+ T cells were reduced in KRAS-mutant CRC compared to wildtype CRC patients. g High SLC25A22 expression correlated with reduced CD8+ T-cell in KRAS-mutant CRC (All cases: n = 202; KRAS-mutant: n = 102). Each dot represents an independent patient. Data are shown as mean ± SD (b, c, d, e, g) and ± SEM for growth curve b. Two-tailed Student’s t test (b, c, d, e, g). Two-tailed two-way ANOVA for growth curves (b). Chi-Square test (g) and Pearson correlation test (d). Source data are provided as a Source Data file.
Fig. 3
Fig. 3. SLC25A22 knockout abrogates secretion of CXCL1 and CXCL3 in vitro and in vivo.
a RNA-seq of SLC25A22 knockout DLD1 cells and gene set enrichment analysis (GSEA) for the identification of common differentially regulated pathways in SLC-KO1 and SLC-KO2 cells (n = 4). b, c GSEA enrichment scores for differentially regulated gene sets unveiled the cytokine-cytokine receptor interaction signaling pathway as the top pathway depleted in SLC25A22 knockout cells. d Inflammatory Response and Autoimmunity PCR array showed that CXCL1, CXCL3 and IL1B were induced in ApcMin/+KrasG12D/+Villin-Cre mice tumors, but were down-regulated by SLC25A22 knockout (FC > 2). e qPCR validated that SLC25A22 knockout inhibited CXCL1/3 mRNA in DLD1, CT26 and Colo26 cells (n = 3). Each dot represents an independent sample. f Antibody array showed SLC25A22 knockout down-regulated cytokine secretion in DLD1 cells. g Densitometry showed CXCL1 and CXCL1/2/3 were top-down-regulated cytokines. h ELISA confirmed SLC25A22 knockout impaired CXCL1/3 secretion in DLD1 (72 h), CT26 (24 h) and Colo26 (24 h) (n = 3). Each dot represents an independent sample. i Detection of CXCL1/3 in serum and tumors of mice implanted with CT26 allografts (left, n = 5) and ApcMin/+KrasG12D/+ organoid allografts (right, n = 10) with or without SLC25A22. Each dot represents an independent mouse. j SLC25A22 mRNA correlates with CXCL1/2/3 mRNA in TCGA CRC (COADREAD) cohort (n = 677). Each dot represents an independent patient. Data are shown as mean ± SD (e, h, i). Two-tailed one-way ANOVA (e, h, i). Two-tailed Student’s t test analysis for two-group comparison i. Pearson correlation test j. Source data are provided as a Source Data file.
Fig. 4
Fig. 4. SLC25A22 knockout abrogates recruitment and activation of MDSC in KRAS-mutant CRC via a CXCL1-CXCR2 axis.
a Flow cytometry validation of isolated mouse MDSC (CD11b+ Gr-1+) and human MDSC (CD11b+CD33+) from the spleens of tumor-implanted syngeneic mice and PBMC humanized mice, respectively. b Conditioned medium from sgControl cells induced MDSC migration, which was impaired by SLC25A22 knockout (n = 4). Each dot represents an independent sample. c siCXCL1, but not siCXCL3, abrogated induction of MDSC migration in sgControl cell conditioned medium (n = 4). Each dot represents an independent sample. d Anti-Cxcl1 neutralizing antibody (0.25 µg/mL) suppressed MDSC migration in sgControl cell conditioned medium; but had no effect on SLC25A22 knockout cells (CT26: n = 3; Colo26: n = 4). Each dot represents an independent sample. e Recombinant Cxcl1 (1 ng/ml) rescued MDSC migration in CT26-Slc-KO and Colo26-Slc-KO conditioned medium (n = 4). Each dot represents an independent sample. f CXCR2 inhibitors SX682 (2 μM) or SB265610 (10 μM) abolished MDSC migration in sgControl cell conditioned medium (n = 4). Each dot represents an independent sample. g Tumoral CXCL1 positively correlated with MDSC infiltration in ApcMin/+KrasG12D/+ organoids (n = 10) in C57BL/6 mice, CT26 allografts (n = 9) in BALB/c mice, and DLD1 xenografts (sgControl, n = 15; SLC-KO1, n = 10) in PBMC humanized mice. Each dot represents an independent mouse. h qPCR of isolated MDSC from CT26 allografts showed that activation markers PD-L1, iNOS, and ARG1 were enhanced in intratumoral MDSC compared to that of splenic MDSC, and was abrogated in SLC25A22 knockout tumors (n = 3). Each dot represents an independent mouse. i Flow cytometry of MDSC from CT26 allografts mice showed that surface PD-L1 protein on MDSC were induced in tumors compared with spleen, which was impaired in SLC25A22 knockout tumors (spleen: n = 3; tumors: n = 10). Each dot represents an independent sample. j SB265610 suppressed the growth of CT26-sgControl allografts (n = 10) and k downregulated MDSC (n = 9), but had no effect on CT26-Slc-KO allografts in BALB/c mice. MDSC positively correlated with tumor weight (n = 36). Each dot represents an independent tumor. Data are shown as mean ± SD. Two-tailed one-way ANOVA (bd, f, hk). Two-tailed Student’s t test analysis for two-group comparison e. Spearman correlation test (g, k). Source data are provided as a Source Data file.
Fig. 5
Fig. 5. SLC25A22-mediated MDSC recruitment represses T-cell proliferation and activation.
a Workflow of T-cell suppression assay. b T-cell suppression assay showed that MDSC isolated from CT26-Slc-KO allografts have attenuated ability to suppress T-cell proliferation (n = 3) and c expression of cytotoxic markers IFN-γ, TNF-α, and Granzyme B (n = 3). Each dot represents an independent sample. d Tumoral MDSC negatively correlated with CD8+ T cells in ApcMin/+KrasG12D/+ allografts (n = 10), CT26 allografts (n = 10), and DLD1 xenografts (sgControl, n = 15; SLC-KO, n = 10). Each dot represents an independent tumor. e In ApcMin/+KrasG12D/+ organoid allografts (n = 10) and f CT26 allografts (n = 10), SLC25A22 knockout increased the tumor-infiltrating CD8+ T cells expressing activation marker (IFN-γ, TNF-α, and Granzyme B). Each dot represents an independent tumor. g CD8+ T cells depletion with anti-CD8 antibody abrogated growth inhibition by SLC25A22 knockout in CT26 allograft (n = 10). Each dot represents an independent mouse (left) or tumor (right). Data are shown as mean ± SD (b, c, eg). Two-tailed one-way ANOVA (b, c). Two-tailed Student’s t test for two-group comparison (e, f). Two-tailed Mann–Whitney U test g. Two-tailed Pearson correlation test d. ns, no significance. Source data are provided as a Source Data file.
Fig. 6
Fig. 6. SLC25A22 drives glutamine-dependent CXCL1 via asparagine.
a In glutamine-depleted (12 h) DLD1 cells, addition of glutamine (72 h) dose-dependently induced CXCL1 mRNA. SLC25A22 knockout selectively inhibited CXCL1 mRNA at high glutamine levels (n = 3). Each dot represents an independent sample. b After glutamine depletion (12 h), glutamine (72 h) dose-dependently induced CXCL1 secretion in DLD1 cells and was impaired by SLC25A22 knockout (n = 3) (left). In glutamine-depleted (12 h) CT26 cells, glutamine (24 h) dose-dependently induced Cxcl1 secretion. SLC25A22 knockout inhibited Cxcl1 secretion at high glutamine (n = 3) (right). Each dot represents an independent sample. c Transaminase inhibition by aminooxyacetate (AOA, 100 μM) inhibited CXCL1 mRNA; whereas blockade of glutamate dehydrogenase 1 (GDH1) by Purpurin (25 μM) and R162 (25 μM) had an opposite effect (n = 3). A similar effect was observed for CXCL1 secretion (n = 3). Each dot represents an independent sample. d Schematic diagram showing the metabolic outputs of glutamine via SLC25A22. e Supplementation of downstream metabolites (2 mM, 24 h) (α-KG, dimethyl α-ketoglutarate; Succ, dimethyl succinate; OAA, dimethyl oxaloacetate; Asp, aspartate; Asn, asparagine) showed that asparagine restored CXCL1 mRNA in SLC25A22 knockout DLD1 cells (n = 3). Aspartate restored CXCL1 mRNA expression in DLD1-SLC-KO cells overexpressing SLC1A3 (n = 3). Each dot represents an independent sample. f Asparagine (2 mM, 24 h) restored CXCL1 secretion in both DLD1 and CT26 cells with knockout of SLC25A22 (n = 3). Aspartate restored CXCL1 secretion in DLD1-SLC-KO cells overexpressing SLC1A3 (n = 3). Each dot represents an independent sample. g After depleting glutamine (12 h), [13C5]-Glutamine stable isotope labeling and LC-MS was performed in DLD1 and CT26 cells with or without SLC25A22 knockout. Total and 13C-labeled asparagine were reduced by SLC25A22 knockout (n = 3). Each dot represents an independent sample. h Treatment with asparagine (2 mM, 24 h) restored the capacity of DLD1-SLC-KO and CT26-Slc-KO cell conditioned medium to promote MDSC migration, without direct effects on MDSC migration (n = 4). i Validation of ASNS knockdown by western blot (left). LC-MS of DLD1 and CT26 cells co-transfected with siASNS and SLC1A3, followed by incubation with 13C4-aspartate (2 mM, 96 h) confirmed the ASNS blockade (n = 4). Each dot represents an independent sample. j LC-MS showed that Asparaginase (ASNase) (24 h) depleted cellular asparagine in DLD1 and CT16 cells (n = 4). Each dot represents an independent sample. k siASNS (upper) or ASNase (lower) reduced CXCL1 mRNA and l secretion in DLD1 and CT26 cells (n = 3). Each dot represents an independent sample. m Conditioned medium from siASNS (upper) or ASNase-treated (lower) DLD1 and CT26 cells had reduced ability to induce MDSC migration. (n = 4). Each dot represents an independent sample. Data are shown as mean ± SD (ac, em) and ± SEM for metabolite curves g. Two-tailed one-way ANOVA (ac, e, gm). Two-tailed Student’s t test f. ns, no significance. Source data are provided as a Source Data file.
Fig. 7
Fig. 7. SLC25A22-asparagine axis is sensed by SRC to activate the ETS2/CXCL1 signaling in KRAS-mutant CRC.
a Phospho-Kinase Array (n = 1 experiment) revealed that asparagine addition (2 mM, 0.5 h, 4 h) in glutamine-low (0.1 M, 12 h) medium activated p-SRC and validation by Western blot (n = 3 independent biological replicates). b SLC25A22 knockout suppressed p-SRC in DLD1 and CT26 cells (n = 2). c Glutamine (1 mM, 12 h) promoted p-SRC in an SLC25A22-dependent manner. Asparagine (2 mM, 12 h) restored p-SRC in SLC25A22 knockout cells (n = 3 independent biological replicates). d Asparagine increased thermal stability (5 min incubation) of recombinant SRC (n = 3 independent biological replicates). e BIAcore analysis of the binding of asparagine to recombinant SRC (n = 2). f Asparagine-induced recombinant SRC phosphorylation and kinase activity (n = 4). Each dot represents an independent sample. g Overlapping of in silico prediction of CXCL1 transcription factors (TFs), SRC-downstream TFs, and SLC25A22 knockout RNA-seq data revealed ETS2 as a putative transcription factor for CXCL1. h SLC25A22 knockout suppressed ETS2 and p-ETS2 expression, and i ETS2 nuclear localization in DLD1 and CT26 cells (n = 2). j Glutamine (1 mM, 12 h) increased ETS2 protein in a SLC25A22-dependent manner. Asparagine (2 mM, 12 h) rescued ETS2 in SLC25A22 knockout cells (n = 2). k ASNS knockdown suppressed ETS2 expression (n = 2). l ETS2 knockdown reduced CXCL1 mRNA and secretion (n = 3). Each dot represents an independent sample. m ETS2 overexpression rescued CXCL1 mRNA and secretion in SLC25A22 knockout cells (n = 3). Each dot represents an independent sample. n In silico identification of ETS2 binding sites on CXCL1 promoter and validation by ChIP-PCR. o Luciferase reporter assay confirmed that ETS2 activated transcription of CXCL1 promoter (n = 3). p SRC inhibitor Bosutinib (24 h) suppressed ETS2, p-ETS2 expression, and CXCL1 secretion in DLD1-sgControl cells, without any effect in SLC25A22 knockout cells (n = 3). Each dot represents an independent sample. Data are shown as mean ± SD (f, l, m, o, p). Two-tailed one-way ANOVA (f, l, p). Two-tailed Student’s t test (m, o). Source data are provided as a Source Data file.
Fig. 8
Fig. 8. SLC25A22 depletion synergized with anti-PD1 therapy in KRAS-mutant CRC.
a SLC25A22 knockout synergized with anti-PD1 in suppressing the growth of CT26 allografts (n = 10) and b MC38-KrasG12V allografts (n = 8). Each dot represents an independent tumor. c CD8+ T-cell and IFN-γ expression was induced by combined SLC25A22 knockout and anti-PD1, whilst MDSC infiltration was reduced by the combination of SLC25A22 knockout plus anti-PD1 therapy in CT26 allografts (n = 5) and d MC38-KrasG12V allografts (n = 8). Each dot represents an independent tumor. e siSlc25a22-encapsulated nanoparticles (VNP-siSLC25A22) synergized with anti-PD1 to suppress MC38-KrasG12V allografts growth and increased CD8+ T-cell activation (n = 10) and f their effect on IFN-γ levels on CD8+ T cells (siNC+IgG and siSLC+IgG: n = 10 per group; siNC+Anti-PD1 and siSLC+ Anti-PD1: n = 9 per group). Each dot represents an independent tumor. g The overall graphical summary of the study (Created with BioRender.com). Data are presented as mean ± SEM for growth curve (a, b, e) and ± SD for others (af). Two-tailed two-way ANOVA for growth curve comparison (a, b, e). Two-tailed Student’s t test for two-group comparison (a, c). Two-tailed Mann–Whitney U test for two-group comparison (b, df). ns, no significance. Source data are provided as a Source Data file.

References

    1. Le DT, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–413. doi: 10.1126/science.aan6733. - DOI - PMC - PubMed
    1. Overman MJ, et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol. 2017;18:1182–1191. doi: 10.1016/S1470-2045(17)30422-9. - DOI - PMC - PubMed
    1. Guinney J, et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015;21:1350–1356. doi: 10.1038/nm.3967. - DOI - PMC - PubMed
    1. Lal N, et al. KRAS mutation and consensus molecular subtypes 2 and 3 are independently associated with reduced immune infiltration and reactivity in colorectal cancer. Clin. Cancer Res. 2018;24:224–233. doi: 10.1158/1078-0432.CCR-17-1090. - DOI - PMC - PubMed
    1. Liao W, et al. KRAS-IRF2 axis drives immune suppression and immune therapy resistance in colorectal cancer. Cancer Cell. 2019;35:559–572 e557. doi: 10.1016/j.ccell.2019.02.008. - DOI - PMC - PubMed

Publication types

Substances