Skip to main content

Advertisement

Log in

Head-to-Head Comparison of CCN4, DNMT3A, PTPN11, and SPARC as Suppressors of Anti-tumor Immunity

  • Original Article
  • Published:
Cellular and Molecular Bioengineering Aims and scope Submit manuscript

Abstract

Purpose

Emergent cancer cells likely secrete factors that inhibit anti-tumor immunity. To identify such factors, we applied a functional assay with proteomics to an immunotherapy resistant syngeneic mouse melanoma model. Four secreted factors were identified that potentially mediate immunosuppression and could become targets for novel immunotherapies. We tested for consistent clinical correlates in existing human data and verified in vivo whether knocking out tumor cell production of these factors improved immune-mediated control of tumor growth.

Methods

Existing human data was analyzed for clinical correlates. A CRISPR/Cas9 approach to generate knockout cell lines and a kinetic analysis leveraging a Markov Chain Monte Carlo (MCMC) approach quantified the various knockouts’ effect on cells’ intrinsic growth rate. Flow cytometry was used to characterize differences in immune infiltration.

Results

While all four gene products were produced by malignant melanocytes, only increased CCN4 expression was associated with reduced survival in primary melanoma patients. In immunocompetent C57BL/6 mice the CCN4 knockout increased survival while the other knockouts had no effect. This survival advantage was lost when the CCN4 knockout cells were injected into immunocompromised hosts, indicating that the effect of CCN4 may be immune mediated. Parameter estimation from the MCMC analysis shows that CCN4 was the only knockout tested that decreased the net tumor growth rate in immunocompetent mice. Flow cytometry showed an increase in NK cell infiltration in CCN4 knockout tumors.

Conclusions

The results suggest that CCN4 is a mediator of immunosuppression in the melanoma tumor microenvironment and a potential collateral immunotherapy target.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Data Availability

Bulk tissue transcriptomics profiling data using Illumina RNA sequencing was accessed from the cutaneous melanoma (SKCM) arm of the Cancer Genome Atlas. Data were downloaded from TCGA data commons using the “TCGAbiolinks” (V2.14.1) package in R (V3.6.3). Gene expression data were expressed in counts. The single cell RNAseq datasets used in the analysis for this article are available in Gene Expression Omnibus repository with the following GEO accession numbers: GSE115978 and GSE72056

Code Availability

The code used in the Markov Chain Monte Carlo analysis can be obtained from the following GitHub repository: https://github.com/arcoolbaugh/B16-In-Vivo-Screen

Abbreviations

IL-12:

Interleukin 12

DNMT3A:

DNA methyltransferase 3

PTPN11:

Protein Tyrosine Phosphatase Non-Receptor Type 11

CCN4:

Cellular Communication Network Factor 4 or Wnt-inducible signaling pathway protein-1

SPARC:

Secreted Protein Acidic and Rich in Cysteine

ATCC:

American Tissue Culture Collection

NSG:

NOD-scid IL2R γnull immunodeficient mice

WVU:

West Virginia University

SKCM:

Skin cutaneous melanoma

TCGA:

The Cancer Genome Atlas

scRNAseq:

Single cell RNA sequencing

MCMC:

Markov Chain Monte Carlo

WT:

Wildtype

KO:

Knock out

References

  1. Tumeh, P. C., et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 515(7528):568–571, 2014. https://doi.org/10.1038/nature13954.

    Article  Google Scholar 

  2. Ganesan, S., and J. Mehnert. Biomarkers for response to immune checkpoint blockade. Annu. Rev. Cancer Biol. 4:331–351, 2020. https://doi.org/10.1146/annurev-cancerbio-030419-033604.

    Article  Google Scholar 

  3. Garris, C. S., et al. Successful anti-PD-1 cancer immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity. 49(6):1148-1161.e7, 2018. https://doi.org/10.1016/j.immuni.2018.09.024.

    Article  Google Scholar 

  4. Kulkarni, Y. M., E. Chambers, A. J. R. McGray, J. S. Ware, J. L. Bramson, and D. J. Klinke. A quantitative systems approach to identify paracrine mechanisms that locally suppress immune response to Interleukin-12 in the B16 melanoma model. Integr. Biol. (United Kingdom). 4(8):925–936, 2012. https://doi.org/10.1039/c2ib20053h.

    Article  Google Scholar 

  5. Wu, Y., W. Deng, E. C. McGinley, and D. J. Klinke. Melanoma exosomes deliver a complex biological payload that upregulates PTPN11 to suppress T lymphocyte function. Pigment Cell Melanoma Res. 30(2):203–218, 2017. https://doi.org/10.1111/pcmr.12564.

    Article  Google Scholar 

  6. Bland, C. L., C. N. Byrne-Hoffman, A. Fernandez, S. L. Rellick, W. Deng, and D. J. Klinke. Exosomes derived from B16F0 melanoma cells alter the transcriptome of cytotoxic T cells that impacts mitochondrial respiration. FEBS J. 285(6):1033–1050, 2018. https://doi.org/10.1111/febs.14396.

    Article  Google Scholar 

  7. Klinke, D. J., A. Fernandez, W. Deng, A. Razazan, H. Latifizadeh, and A. C. Pirkey. Data-driven learning how oncogenic gene expression locally alters heterocellular networks. Nat. Commun. 13(1):1986, 2022. https://doi.org/10.1038/s41467-022-29636-3.

    Article  Google Scholar 

  8. Deng, W., A. Fernandez, S. L. McLaughlin, and D. J. Klinke. WNT1-inducible signaling pathway protein 1 (WISP1/CCN4) stimulates melanoma invasion and metastasis by promoting the epithelial mesenchymal transition. J. Biol. Chem. 294(14):5261–5280, 2019. https://doi.org/10.1074/jbc.RA118.006122.

    Article  Google Scholar 

  9. Jerby-Arnon, L., et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell. 175(4):984-997.e24, 2018. https://doi.org/10.1016/j.cell.2018.09.006.

    Article  Google Scholar 

  10. Tirosh, I., et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science (1979). 352(6282):189–196, 2016. https://doi.org/10.1126/science.aad0501.

    Article  Google Scholar 

  11. Klinke, D. J. In silico model-based inference: a contemporary approach for hypothesis testing in network biology. Biotechnol. Prog. 30(6):1247–1261, 2014. https://doi.org/10.1002/btpr.1982.

    Article  Google Scholar 

  12. Deng, W., A. Fernandez, S. L. McLaughlin, and D. J. Klinke. Cell Communication Network Factor 4 (CCN4/WISP1) shifts melanoma cells from a fragile proliferative state to a resilient metastatic state. Cell Mol. Bioeng. 13(1):45–60, 2020. https://doi.org/10.1007/s12195-019-00602-2.

    Article  Google Scholar 

  13. Overwijk, W. W., and N. P. Restifo. B16 as a mouse model for human melanoma. Curr. Protoc. Immunol. 2000. https://doi.org/10.1002/0471142735.im2001s39.

    Article  Google Scholar 

  14. Byrne-Hoffman, C. N., W. Deng, O. McGrath, P. Wang, Y. Rojanasakul, and D. J. Klinke. Interleukin-12 elicits a non-canonical response in B16 melanoma cells to enhance survival. Cell Commun. Signal. 2020. https://doi.org/10.1186/s12964-020-00547-4.

    Article  Google Scholar 

  15. Jia, H., et al. The tumor cell-secreted matricellular protein WISP 1 drives pro-metastatic collagen linearization. EMBO J. 38(16):1–15, 2019. https://doi.org/10.15252/embj.2018101302.

    Article  Google Scholar 

  16. Wang, Q. Y., Y. J. Feng, and R. Ji. High expression of WISP1 promotes metastasis and predicts poor prognosis in hepatocellular carcinoma. Eur. Rev. Med. Pharmacol. Sci. 24(20):10445–10451, 2020. https://doi.org/10.26355/eurrev_202010_23396.

    Article  Google Scholar 

  17. Fernandez, A., et al. Cell communication network factor 4 promotes tumor-induced immunosuppression in melanoma. EMBO Rep. 2022. https://doi.org/10.15252/embr.202154127.

    Article  Google Scholar 

  18. Hou, C. H., C. H. Tang, C. J. Hsu, S. M. Hou, and J. F. Liu. CCN4 induces IL-6 production through αvβ5 receptor, PI3K, Akt, and NF-κB singling pathway in human synovial fibroblasts. Arthritis Res. Ther. 15(1):R19, 2013. https://doi.org/10.1186/ar4151.

    Article  Google Scholar 

  19. Ono, M., et al. CCN4/WISP1 controls cutaneous wound healing by modulating proliferation, migration and ECM expression in dermal fibroblasts via α5β1 and TNFα. Matrix Biol. 68–69:533–546, 2018. https://doi.org/10.1016/j.matbio.2018.01.004.

    Article  Google Scholar 

  20. Tao, W., et al. Dual Role of WISP1 in maintaining glioma stem cells and tumor-supportive macrophages in glioblastoma. Nat. Commun. 2020. https://doi.org/10.1038/s41467-020-16827-z.

    Article  Google Scholar 

  21. Wu, S. Z., et al. A single-cell and spatially resolved atlas of human breast cancers. Nat. Genet. 53(9):1334–1347, 2021. https://doi.org/10.1038/s41588-021-00911-1.

    Article  Google Scholar 

  22. Chen, B., and W. Chan. The de novo DNA methyltransferase DNMT3A in development and cancer. Epigenetics. 9(5):669–677, 2014. https://doi.org/10.4161/epi.28324.

    Article  Google Scholar 

  23. Robertson, K. D., et al. The human DNA methyltransferases (DNMTs) 1, 3a and 3b: coordinate mRNA expression in normal tissues and overexpression in tumors. Nucleic Acids Res. 27(11):2291–2298, 1999. https://doi.org/10.1093/nar/27.11.2291.

    Article  Google Scholar 

  24. Zhang, W., and J. Xu. DNA methyltransferases and their roles in tumorigenesis. Biomark. Res. 5(1):1–8, 2017. https://doi.org/10.1186/s40364-017-0081-z.

    Article  Google Scholar 

  25. Deng, T., Y. Kuang, L. Wang, J. Li, Z. Wang, and J. Fei. An essential role for DNA methyltransferase 3a in melanoma tumorigenesis. Biochem. Biophys. Res. Commun. 387(3):611–616, 2009. https://doi.org/10.1016/j.bbrc.2009.07.093.

    Article  Google Scholar 

  26. Moufarrij, S., et al. Combining DNMT and HDAC6 inhibitors increases anti-tumor immune signaling and decreases tumor burden in ovarian cancer. Sci. Rep. 2020. https://doi.org/10.1038/s41598-020-60409-4.

    Article  Google Scholar 

  27. Segovia, C., et al. Inhibition of a G9a/DNMT network triggers immune-mediated bladder cancer regression. Nat. Med. 25(7):1073–1081, 2019. https://doi.org/10.1038/s41591-019-0499-y.

    Article  Google Scholar 

  28. Hill, K. S., et al. PTPN11 plays oncogenic roles and is a therapeutic target for BRAF wild-type melanomas. Mol. Cancer Res. 17(2):583–593, 2019. https://doi.org/10.1158/1541-7786.MCR-18-0777.

    Article  Google Scholar 

  29. Girotti, M. R., et al. SPARC promotes cathepsin B-mediated melanoma invasiveness through a collagen i/α2Β1 integrin axis. J. Invest. Dermatol. 131(12):2438–2447, 2011. https://doi.org/10.1038/jid.2011.239.

    Article  Google Scholar 

  30. Jiang, Y., and H. Zhan. Communication between EMT and PD-L1 signaling: new insights into tumor immune evasion. Cancer Lett. 468(2019):72–81, 2020. https://doi.org/10.1016/j.canlet.2019.10.013.

    Article  Google Scholar 

  31. Haber, C. L., et al. SPARC modulates the proliferation of stromal but not melanoma cells unless endogenous SPARC expression is downregulated. Int. J. Cancer. 122(7):1465–1475, 2008. https://doi.org/10.1002/ijc.23216.

    Article  Google Scholar 

  32. Prada, F., L. G. Benedetti, A. I. Bravo, M. J. Alvarez, C. Carbone, and O. L. Podhajcer. SPARC endogenous level, rather than fibroblast-produced SPARC or stroma reorganization induced by SPARC, is responsible for melanoma cell growth. J. Invest. Dermatol. 127(11):2618–2628, 2007. https://doi.org/10.1038/sj.jid.5700962.

    Article  Google Scholar 

  33. Manguso, R. T., et al. In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. Nature. 547(7664):413–418, 2017. https://doi.org/10.1038/nature23270.

    Article  Google Scholar 

  34. Masoudi, M., M. Seki, R. Yazdanparast, N. Yachie, and H. Aburatani. A genome-scale CRISPR/Cas9 knockout screening reveals SH3D21 as a sensitizer for gemcitabine. Sci. Rep. 9(1):1–9, 2019. https://doi.org/10.1038/s41598-019-55893-2.

    Article  Google Scholar 

Download references

Acknowledgements

Figure 4 was created with Biorender.com, for which the subscription was provided by the Cellular and Molecular Biology and Biomedical Engineering NIH T32 Program (GM133369) at West Virginia University. We would also like to thank Audry Fernandez for her assistance with flow cytometry experiments. This work was supported by grants received by David. J. Klinke II from the National Science Foundation (NSF CBET-1644932) and National Cancer Institute (NCI 1R01CA193473). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NSF or NCI. We also used equipment from the WVU Flow Cytometry & Single Cell Core Facility (RRID: SCR_017738). The core was supported by National Institute of Health Grants GM121322, GM103434, OD016165, and GM104942.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to David J. Klinke II.

Ethics declarations

Conflict of interest

The authors declare that they have no conflicts of interest.

Animal Studies

All procedures involving animals were approved by the West Virginia University (WVU) Institutional Animal Care and Use Committee and performed at the WVU Animal Facility ((IACUC Protocol #1604002138).

Additional information

Associate Editor Matthew Lazzara oversaw the review of this article.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

12195_2023_787_MOESM1_ESM.tif

Supplementary file1 (TIF 17743 kb) Supplemental Figure S1: Western blot images confirming the knockout of (A) DNMT3A, (B) PTPN11, (C) SPARC, and (D) CCN4 in B16F0 cells.

12195_2023_787_MOESM2_ESM.tif

Supplementary file2 (TIF 13841 kb) Supplemental Figure S2: Representative images of diagnostics for Markov Chain Monte Carlo estimates of the posterior distribution of initial tumor bolus size in a single mouse. (A) The posterior distribution of the parameter for each of the three independent chains overlap, indicating agreement between the chains. (B) New steps in the Markov Chain were proposed with increased or decreased risk to achieve a desired acceptance fraction of test point of 0.20. (C) The full-length traces of three independent chains (colored in black, blue, and red, respectively) with over disperse starting points. (D) The Gelman-Rubin potential scale reduction factor (PSRF) was used to assess convergence of the Markov Chains to the posterior distribution in each parameter where a value of less than 1.2 indicated that the chains have converged.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pirkey, A.C., Deng, W., Norman, D. et al. Head-to-Head Comparison of CCN4, DNMT3A, PTPN11, and SPARC as Suppressors of Anti-tumor Immunity. Cel. Mol. Bioeng. 16, 431–442 (2023). https://doi.org/10.1007/s12195-023-00787-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12195-023-00787-7

Keywords

Navigation