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
. 2025 Jul 22;11(1):337.
doi: 10.1038/s41420-025-02627-1.

MARCO expression on myeloid-derived suppressor cells is essential for their differentiation and immunosuppression

Affiliations

MARCO expression on myeloid-derived suppressor cells is essential for their differentiation and immunosuppression

Sijia Liu et al. Cell Death Discov. .

Abstract

Myeloid-derived suppressor cells (MDSCs) significantly contribute to the immunosuppressive tumor microenvironment (TME), and targeted inhibition of MDSCs is a potential therapeutic strategy against cancer. Here, we identify macrophage receptor with collagenous structure (MARCO) as a critical regulator of MDSC differentiation and immunosuppression in breast cancer. The present study demonstrates that MARCO is expressed on MDSCs, and breast tumor-derived exosomes (TDEs) enriched with macrophage migration inhibitory factor (MIF) promote MDSC differentiation and amplify immunosuppressive activity by up-regulating MARCO. Genetic ablation of MARCO in a murine breast cancer model attenuated tumor growth, accompanied by reduced monocytic MDSCs (M-MDSCs) and total tumor-associated macrophages (TAMs), along with enhanced infiltration of CD8+ T cells and natural killer (NK) cells. Furthermore, we developed a specific MARCO down-regulation-promoting monoclonal antibody that impeded TDE-induced MDSC differentiation and immunosuppression. In vivo, MARCO down-regulating antibody suppressed tumor growth and reprogrammed the TME by diminishing immunosuppressive MDSCs and TAMs and revitalizing CD8+ T cells and NK cells. Strikingly, combining the MARCO down-regulating antibody with PD-1 blockade synergistically enhanced anti-tumor efficacy. This work establishes MARCO as a key regulator of MDSC-mediated immunosuppression and presents a compelling case for the inclusion of MARCO as a therapeutic target in cancer immunotherapy.

PubMed Disclaimer

Conflict of interest statement

Competing interests: The authors declare no competing interests.

Figures

Fig. 1
Fig. 1. TDE induces MARCO expression and promotes MDSC differentiation.
A Flow cytometric analysis of MARCO, Arg-1, and CD206 expression on cytokine-induced murine bone marrow cells. (M1: M-CSF + LPS + IFN-γ; M2: M-CSF + IL4 + IL10; MDSC: GM-CSF + IL6). BD MDSC proportion, MDSC MARCO expression, and MDSC Arg-1 expression on murine bone marrow cells treated with E0771 tumor-sup or TDEs. E M-MDSC and G-MDSC proportion. Murine BM cells treated with E0771 tumor-sup or TDE. F Representative flow cytometry figure of MDSC subgroups. G Human PBMC cells were treated with different concentrations of MDA-MMB-231 TDE. Then, the MDSC proportion was detected. H Representative flow cytometry figure of MDSC proportion. I MARCO expression on human TDE-induced MDSC. Data are presented as mean ± SD with a CI of 95% from three biological replicates (individual data points overlaid), and the entire study was independently repeated at least three times. p < 0.05 by one-way ANOVA with Tukey’s multiple comparisons test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.001.
Fig. 2
Fig. 2. MIF in TDE promotes MDSC differentiation and MARCO upregulation.
A MIF RNA expression of MDA-MBA-231 cells transfected with different indicated plasmids. B Representative Western blot images demonstrate the expression of MIF in TDEs isolated from MDA-MB-231 transfected with MIF shRNA (sh-MIF) or negative control shRNA (NC). Three biological replicates per group. C Relative gray value analysis of Western blot images to prove MIF knockdown in exosomes. D The proportions of MDSCs when human PBMCs respond to the sh-MIF or NC TDEs. Each experiment included four biological replicates, and the entire study was independently repeated at least three times. E Representative flow cytometry figure of (D). F The proportions of human PBMC differentiated M-MDSCs and MARCO+ MDSC in response to the sh-MIF or NC TDEs. G Representative flow cytometry figure of (F). H Mouse BM cells induced by E0771 exosomes were treated with IMP-1576 with different concentrations, compared to BM without E0771 TDE. Quantitation data of M-MDSC and MARCO+ MDSC proportion. I Representative flow cytometry figure of (H). Data was shown as mean ± SD. with a CI of 95% and individual data points overlaid. p < 0.05 by one-way ANOVA with Tukey’s multiple comparisons test for pairwise comparisons between groups. **p < 0.01, ***p < 0.001, ****p < 0.001. ns: no significance.
Fig. 3
Fig. 3. MARCO deficiency prevented tumor growth and altered the characteristics of MDSCs.
A Schematic of the E0771 mammary tumor model and period of tumor volume measurement. (n = 4 for both wild-type and MARCO Knockout groups littermate controls). B The growth curve of E0771 breast tumors. Data was shown as mean ± SEM. with a CI of 95%. We analyzed tumor growth differences between the two groups across multiple time points using two-way ANOVA (factors: Group × Time) with Šídák’s multiple comparisons test to show the difference between groups at the same time point. C The tumor weight at the experimental endpoint. D, E Proportions and representative flow cytometry data of G-MDSCs and M-MDSCs in tumor tissue. F, G Proportions and representative flow cytometry data of TAM. H, I Quantification and representative flow cytometry data of tumor-infiltrating CD8+ T cells. J, K Quantification and representative flow cytometry data of tumor-infiltrating NK cells. Data in (CK) was shown as mean ± SD. with a CI of 95%. Comparisons between the two groups were performed using unpaired two-tailed Student’s t-tests. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.001, ns not significant. L MDSCs were isolated from the MARCO deficiency and wild-type group (n = 3 for each group) for RNA sequencing analysis. The heatmap showed the differentially expressed genes between knockout MDSCs and wild-type MDSCs.
Fig. 4
Fig. 4. MARCO downregulation-inducing antibody inhibits the MDSC differentiation.
Mouse bone marrow cells incubated with E0771 TDEs in the presence of control antibody (CTRL) or MARCO antibody (2L4-8) 30 μg/mL. A flow cytometry to detect MARCO and Arg-1 expression on MDSCs. B RT-PCR to detect total MARCO, PD-L1, and Arg-1 expression. The experiment contained four biological replicates and was independently repeated at least three times. C, D Representative flow cytometry data and quantification analysis of the proportion of MDSCs’ sub-populations. E, F TDE-induced MDSCs were co-cultured with CD8+ T cells at a 1:1 ratio with the presence of isotype control and 2L4-8 antibodies for 2 days. Three biological replicates per group. Representative flow cytometry data showed the proliferation of CD8+ T cells. Quantification of CD8+ T cell proliferation. G Quantification data showed the dose-dependent effect of 2L4-8 to inhibit the MARCO and CD74 expression on M-MDSCs. The experiment contained four biological replicates and was independently repeated at least three times. H Representative flow cytometry of (G). Data were shown as mean ± SD. with a CI of 95%. For experiments with three groups, One-way ANOVA was performed to assess overall differences among the three groups, followed by Tukey’s post hoc test for all pairwise comparisons. For experiments with two groups, an unpaired two-tailed Student’s t-test was used. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Fig. 5
Fig. 5. MARCO downregulation antibody attenuates TDE-induced MDSC immunosuppression.
A Experimental scheme of RNA-seq. Control vs. antibody-treated group (n = 4 for each group). B Volcano plot displaying differentially expressed genes (DEGs) between treatment and control groups (n = 4 each group). The X-axis shows log2 fold change, where positive values indicate higher expression in the treatment group. The Y-axis represents –log10 adjusted p-value. Red dots denote significant DEGs (|LFC | > 2, padj < 0.0001). C Data from GO enrichment analysis (GO:BP) shows the genes enriched in signaling pathways. D GSEA analysis showed the signaling pathway change in the two groups. The results are shown in a bar graph. E Heatmap of essential downregulating genes in response to 2L4-8 treatment. F All sequenced samples were subjected to RT-PCR to validate the expression of MARCO, Csf1, IL10, Il1a, Il1b, Cxcl14, and Fosl1. RT-PCR Data was shown as mean ± SD. with a CI of 95%. An unpaired two-tailed Student’s t-test was used. **p < 0.01, ***p < 0.001, ****p < 0.0001.
Fig. 6
Fig. 6. The MARCO antibody 2L4-8 enhanced the anti-tumor immune response of PD-1 therapy.
A Schematic of the E0771 mammary tumor model and treatment regimen from day 9 to 21 after inoculation. N = 5 for each group. B E0771 tumor growth in response to different treatments. Data was shown as mean ± SEM with a CI of 95%. The tumor growth differences between the two groups across multiple time points were analyzed by two-way ANOVA (factors: Group × Time) with Šídák’s multiple comparisons test to show the difference between groups at the same time point. C Tumor weight of the E0771 tumor. D Proportions of tumor-infiltrating G-MDSCs and M-MDSCs in each group. E Representative flow cytometry data of MDSC subtypes. F Proportion of MARCO+ M-MDSC and in Arg-1+ M-MDSC each group. G Percentage of tumor-infiltrating TAM in each group. K Representative flow cytometry data of TAMs. H Representative flow cytometry data of TAMs. I Percentage of tumor-infiltrating CD206+TAM and CD86+ TAM in each group. J, K Percentages of tumor-infiltrating CD8+ T cells and PD-1+ CD8+ T cells in each group. L, M Percentages of tumor-infiltrating NK cells and IFN-γ+ NK cells in each group. The weight of the tumor and flow cytometry data was analyzed using one-way ANOVA with Tukey’s multiple comparisons test for pairwise comparisons between groups. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.001. ns: no significance.

Similar articles

References

    1. Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat Rev Cancer. 2003;3:453–8. 10.1038/nrc1098. - PubMed
    1. de Visser KE, Joyce JA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell. 2023;41:374–403. 10.1016/j.ccell.2023.02.016. - PubMed
    1. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–37. 10.1038/nm.3394. - PMC - PubMed
    1. Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC, et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol. 2001;166:678–89. 10.4049/jimmunol.166.1.678. - PubMed
    1. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162–74. 10.1038/nri2506. - PMC - PubMed

LinkOut - more resources