Skip to main content
Log in

Exosomal microRNA-4535 of Melanoma Stem Cells Promotes Metastasis by Inhibiting Autophagy Pathway

  • Published:
Stem Cell Reviews and Reports Aims and scope Submit manuscript

Abstract

High mortality rate and poor survival in melanoma are associated with efficient metastatic colonization. The underlying mechanisms remain elusive. Elucidating the role of exosomes in mediating the interactions between cancer cells and the metastatic microenvironment has been focused on cancer cell derived exosomes in modulating the functions of stromal cells. Whether cancer stem cells (CSCs) can modify the metastatic properties of non-CSC cells, and whether exosomal crosstalk plays a role have not been demonstrated prior to this report. In this study, a paired M14 melanoma derivative cell line, i.e., melanoma parental cell (MPC) and its CSC derivative cell line melanoma stem cell (MSC) were employed. We demonstrated that exosomal crosstalk betwen MSCs and non-CSC MPCs is a new mechanism that underlies melanoma metastasis. Low metastatic melanoma cells (MPCs) can acquire the “metastatic power” from highly metastatic melanoma CSCs (MSCs). We illustrated an uncharacterized microRNA, miR-4535 in mediating such exosomal crosstalk. MSCs deliver its exosomal miR-4535 to the targeted MPCs. Upon entering MPCs, miR-4535 augments metastatic colonization of MPCs by inactivating the autophagy pathway.

Graphical Abstract

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

Similar content being viewed by others

Data Availability

The data used to support the findings of this study are available from every author upon request.

References

  1. Gensbittel, V., et al. (2021). Mechanical adaptability of tumor cells in metastasis. Developmental Cell, 56(2), 164–179.

    Article  CAS  Google Scholar 

  2. Schadendorf, D., et al. (2018). Melanoma. The Lancet, 392(10151), 971–984.

    Article  Google Scholar 

  3. Batlle, E., & Clevers, H. (2017). Cancer stem cells revisited. Nature Medicine, 23(10), 1124–1134.

    Article  CAS  Google Scholar 

  4. Regan, J. L., et al. (2017). Non-canonical hedgehog signaling is a positive regulator of the WNT pathway and is required for the survival of Colon Cancer stem cells. Cell Reports, 21(10), 2813–2828.

    Article  CAS  Google Scholar 

  5. Lee, S. Y., et al. (2017). Induction of metastasis, cancer stem cell phenotype, and oncogenic metabolism in cancer cells by ionizing radiation. Molecular Cancer, 16(1), 1–25.

    Article  Google Scholar 

  6. Hu, J., et al. (2017). A CD44v(+) subpopulation of breast cancer stem-like cells with enhanced lung metastasis capacity. Cell Death & Disease, 8(3), 1–9.

    Article  Google Scholar 

  7. Lawson, D. A., et al. (2015). Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature, 526(7571), 131–135.

    Article  CAS  Google Scholar 

  8. Luzzi, K. J., et al. (1998). Dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Multistep Nature of Metastatic Inefficiency, 153, 865–873.

    CAS  Google Scholar 

  9. Chen, D., et al. (2016). The neuropeptides FLP-2 and PDF-1 act in concert to arouse Caenorhabditis elegans locomotion. Genetics, 204(3), 1151–1159.

    Article  CAS  Google Scholar 

  10. Barnes, D. G., et al. (2013). Embedding and publishing interactive, 3-dimensional, scientific figures in portable document format (PDF) files. PLoS One, 8(9), 1559–1564.

    Article  Google Scholar 

  11. Lenos, K. J., et al. (2018). Stem cell functionality is microenvironmentally defined during tumour expansion and therapy response in colon cancer. Nature Cell Biology, 20(10), 1193–1202.

    Article  CAS  Google Scholar 

  12. Li, F., et al. (2019). Retinoblastoma inactivation induces a Protumoral microenvironment via enhanced CCL2 secretion. Cancer Research, 79(15), 3903–3915.

    Article  CAS  Google Scholar 

  13. Wilson, J. L., et al. (2020). Inverse data-driven modeling and multiomics analysis reveals Phgdh as a metabolic checkpoint of macrophage polarization and proliferation. Cell Reports, 30(5), 1542–1552.

    Article  CAS  Google Scholar 

  14. Plaks, V., et al. (2015). The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell, 16(3), 225–238.

    Article  CAS  Google Scholar 

  15. Wortzel, I., et al. (2019). Exosome-mediated metastasis: Communication from a distance. Developmental Cell, 49(3), 347–360.

    Article  CAS  Google Scholar 

  16. Devhare, P. B., & Ray, R. B. (2018). Extracellular vesicles: Novel mediator for cell to cell communications in liver pathogenesis. Molecular Aspects of Medicine, 60, 115–122.

    Article  CAS  Google Scholar 

  17. Rauner, G., et al. (2018). High expression of CD200 and CD200R1 distinguishes stem and progenitor cell populations within mammary repopulating units. Stem Cell Reports, 11(1), 288–302.

    Article  CAS  Google Scholar 

  18. Mathieu, M., et al. (2019). Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nature Cell Biology, 21(1), 9–17.

    Article  CAS  Google Scholar 

  19. Bonsergent, E., et al. (2021). Quantitative characterization of extracellular vesicle uptake and content delivery within mammalian cells. Nature Communications, 12(1), 1–11.

    Article  Google Scholar 

  20. Ratajczak, M. Z., & Ratajczak, J. (2020). Extracellular microvesicles/exosomes: Discovery, disbelief, acceptance, and the future? Leukemia, 34(12), 3126–3135.

    Article  Google Scholar 

  21. Pegtel, D. M., & Gould, S. J. (2019). Exosomes. Annual Review of Biochemistry, 88, 487–514.

    Article  CAS  Google Scholar 

  22. Spilak, A., et al. (2021). Implications and pitfalls for cancer diagnostics exploiting extracellular vesicles. Advanced Drug Delivery Reviews, 175, 1–18.

    Article  Google Scholar 

  23. Kalluri, R., & LeBleu, V. S. (2020). The biology, function, and biomedical applications of exosomes. science, 367, 1–15.

    Article  Google Scholar 

  24. Galasso, M., et al. (2012). MicroRNA expression signatures in solid malignancies. The Cancer Journal, 18, 238–243.

    Article  CAS  Google Scholar 

  25. Xie, Y., et al. (2019). The role of exosomal noncoding RNAs in cancer. Molecular Cancer, 18(1), 1–10.

    Article  CAS  Google Scholar 

  26. Pomatto, M. A. C., et al. (2019). Improved loading of plasma-derived extracellular vesicles to encapsulate antitumor miRNAs. Mol Ther Methods Clin Dev, 13, 133–144.

    Article  CAS  Google Scholar 

  27. Sun, Z., et al. (2018). Effect of exosomal miRNA on cancer biology and clinical applications. Molecular Cancer, 17(1), 1–19.

    Article  Google Scholar 

  28. Zhang, L., & Yu, D. (2019). Exosomes in cancer development, metastasis, and immunity. Biochimica Et Biophysica Acta. Reviews on Cancer, 1871(2), 455–468.

    Article  CAS  Google Scholar 

  29. Cavallari, C., et al. (2020). Extracellular vesicles in the tumour microenvironment: Eclectic supervisors. International Journal of Molecular Sciences, 21(18), 1–21.

    Article  Google Scholar 

  30. Zeng, Z., et al. (2018). Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nature Communications, 9(1), 1–14.

    Article  Google Scholar 

  31. Fang, T., et al. (2018). Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nature Communications, 9(1), 1–13.

    Article  Google Scholar 

  32. Yu, L., et al. (2021). Exosomes derived from osteogenic tumor activate osteoclast differentiation and concurrently inhibit osteogenesis by transferring COL1A1-targeting miRNA-92a-1-5p. J Extracell Vesicles, 10(3), 1–27.

    Article  Google Scholar 

  33. Zhao, S., et al. (2020). Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. Journal of Hematology & Oncology, 13(1), 1–19.

    Article  Google Scholar 

  34. Lopatina, T., et al. (2020). Targeting IL-3Ralpha on tumor-derived endothelial cells blunts metastatic spread of triple-negative breast cancer via extracellular vesicle reprogramming. Oncogenesis, 9(10), 1–14.

    Article  Google Scholar 

  35. Lopatina, T., et al. (2020). Extracellular vesicles released by tumor endothelial cells spread immunosuppressive and transforming signals through various recipient cells. Frontiers in Cell and Development Biology, 8, 1–14.

    Article  Google Scholar 

  36. Lombardo, G., et al. (2018). IL-3R-alpha blockade inhibits tumor endothelial cell-derived extracellular vesicle (EV)-mediated vessel formation by targeting the beta-catenin pathway. Oncogene, 37(9), 1175–1191.

    Article  CAS  Google Scholar 

  37. Yang, B., et al. (2020). High-metastatic cancer cells derived exosomal miR92a-3p promotes epithelial-mesenchymal transition and metastasis of low-metastatic cancer cells by regulating PTEN/Akt pathway in hepatocellular carcinoma. Oncogene, 39(42), 6529–6543.

    Article  CAS  Google Scholar 

  38. Wang, L., et al. (2019). CD103-positive CSC exosome promotes EMT of clear cell renal cell carcinoma: Role of remote MiR-19b-3p. Molecular Cancer, 18(1), 1–15.

    Article  Google Scholar 

  39. Wang, Z. F., et al. (2019). Glioma stem cells-derived exosomal miR-26a promotes angiogenesis of microvessel endothelial cells in glioma. Journal of Experimental & Clinical Cancer Research, 38(1), 1–15.

    Google Scholar 

  40. Lopez de Andres, J., et al. (2020). Cancer stem cell secretome in the tumor microenvironment: A key point for an effective personalized cancer treatment. Journal of Hematology & Oncology, 13(1), 1–22.

    Article  Google Scholar 

  41. Somasundaram, R., & Herlyn, M. (2012). Melanoma exosomes: messengers of metastasis. Nature Medicine, 18(6), 853–854.

    Article  CAS  Google Scholar 

  42. Kim, J., et al. (2018). Replication study: Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Elife, 7, 1–17.

    Article  Google Scholar 

  43. Vignard, V., et al. (2020). MicroRNAs in tumor exosomes drive immune escape in melanoma. Cancer Immunology Research, 8(2), 255–267.

    Article  CAS  Google Scholar 

  44. Wang, J., et al. (2017). Comparison of tumor biology of two distinct cell sub-populations in lung cancer stem cells. Oncotarget, 8(57), 96852–96864.

    Article  Google Scholar 

  45. Sun, Z., et al. (2018). Sec23a mediates miR-200c augmented oligometastatic to polymetastatic progression. EBioMedicine, 37, 47–55.

    Article  Google Scholar 

  46. Song, L., et al. (2019). KIBRA controls exosome secretion via inhibiting the proteasomal degradation of Rab27a. Nature Communications, 10(1), 1–13.

    Article  Google Scholar 

  47. Makarova, J., et al. (2021). Extracellular miRNAs and cell-cell communication: Problems and prospects. Trends in Biochemical Sciences, 46(8), 640–651.

    Article  CAS  Google Scholar 

  48. Wu, K., et al. (2021). Exosomal miR-19a and IBSP cooperate to induce osteolytic bone metastasis of estrogen receptor-positive breast cancer. Nature Communications, 12(1), 1–18.

    Google Scholar 

  49. Fang, J. H., et al. (2018). Hepatoma cell-secreted exosomal microRNA-103 increases vascular permeability and promotes metastasis by targeting junction proteins. Hepatology, 68(4), 1459–1475.

    Article  CAS  Google Scholar 

  50. Liu, C., et al. (2017). MicroRNA-141 suppresses prostate cancer stem cells and metastasis by targeting a cohort of pro-metastasis genes. Nature Communications, 8, 1–14.

    Google Scholar 

  51. Barkan, D., et al. (2008). Inhibition of metastatic outgrowth from single dormant tumor cells by targeting the cytoskeleton. Cancer Research, 68(15), 6241–6250.

    Article  CAS  Google Scholar 

  52. Sun, Z., et al. (2020). S100A8 transported by SEC23A inhibits metastatic colonization via autocrine activation of autophagy. Cell Death & Disease, 11(8), 1–13.

    Article  Google Scholar 

  53. Ono, M., et al. (2014). Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in metastatic breast cancer cells. Science Signaling, 7(332), 1–10.

    Article  Google Scholar 

  54. Temoche-Diaz, M. M., et al. (2019). Distinct mechanisms of microRNA sorting into cancer cell-derived extracellular vesicle subtypes. Elife, 8, 1–34.

    Article  Google Scholar 

  55. Grzywa, T. M., et al. (2017). Intratumor and Intertumor heterogeneity in melanoma. Translational Oncology, 10(6), 956–975.

    Article  Google Scholar 

  56. Zhang, X., et al. (2015). Exosomes in cancer: Small particle, big player. Journal of Hematology & Oncology, 8, 1–13.

    Article  Google Scholar 

  57. Wang, L., et al. (2020). Lung CSC-derived exosomal miR-210-3p contributes to a pro-metastatic phenotype in lung cancer by targeting FGFRL1. Journal of Cellular and Molecular Medicine, 24(11), 6324–6339.

    Article  CAS  Google Scholar 

  58. Li, J., et al. (2021). Hypoxic glioma stem cell-derived exosomes containing Linc01060 promote progression of glioma by regulating the MZF1/c-Myc/HIF1alpha Axis. Cancer Research, 81(1), 114–128.

    Article  CAS  Google Scholar 

  59. Hardin, H., et al. (2018). Thyroid cancer stem-like cell exosomes: Regulation of EMT via transfer of lncRNAs. Laboratory Investigation, 98(9), 1133–1142.

    Article  CAS  Google Scholar 

  60. Yang, Z., et al. (2020). Exosomes derived from cancer stem cells of gemcitabine-resistant pancreatic cancer cells enhance drug resistance by delivering miR-210. Cellular Oncology (Dordrecht), 43(1), 123–136.

    Article  CAS  Google Scholar 

  61. Hwang, W. L., et al. (2019). Tumor stem-like cell-derived exosomal RNAs prime neutrophils for facilitating tumorigenesis of colon cancer. Journal of Hematology & Oncology, 12(1), 1–17.

    Article  Google Scholar 

  62. Naseri, M., et al. (2021). Dendritic cells loaded with exosomes derived from cancer stem cell-enriched spheroids as a potential immunotherapeutic option. Journal of Cellular and Molecular Medicine, 25(7), 3312–3326.

    Article  CAS  Google Scholar 

  63. Yuan, Y., et al. (2021). Exosomal O-GlcNAc transferase from esophageal carcinoma stem cell promotes cancer immunosuppression through up-regulation of PD-1 in CD8(+) T cells. Cancer Letters, 500, 98–106.

    Article  CAS  Google Scholar 

  64. Sun, Z., et al. (2020). Glioblastoma stem cell-derived exosomes enhance Stemness and Tumorigenicity of glioma cells by transferring Notch1 protein. Cellular and Molecular Neurobiology, 40(5), 767–784.

    Article  CAS  Google Scholar 

  65. Dai, W., et al. (2020). Exosomal lncRNA DOCK9-AS2 derived from cancer stem cell-like cells activated Wnt/beta-catenin pathway to aggravate stemness, proliferation, migration, and invasion in papillary thyroid carcinoma. Cell Death & Disease, 11(9), 1–17.

    Article  Google Scholar 

  66. Cha, S. Y., et al. (2017). Clinical impact of microRNAs associated with Cancer stem cells as a prognostic factor in ovarian carcinoma. Journal of Cancer, 8(17), 3538–3547.

    Article  Google Scholar 

  67. Conigliaro, A., et al. (2015). CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Molecular Cancer, 14, 1–11.

    Article  Google Scholar 

  68. Yoshikawa, K., et al. (2021). Diagnostic predictability of miR-4535 and miR-1915-5p expression in amniotic fluid for foetal morbidity of infection. Placenta, 114, 68–75.

    Article  CAS  Google Scholar 

  69. Hannafon, B. N., et al. (2016). Plasma exosome microRNAs are indicative of breast cancer. Breast Cancer Research, 18(1), 1–14.

    Article  Google Scholar 

  70. Hydbring, P., et al. (2018). Exosomal RNA-profiling of pleural effusions identifies adenocarcinoma patients through elevated miR-200 and LCN2 expression. Lung Cancer, 124, 45–52.

    Article  Google Scholar 

  71. Lee, C. H., et al. (2018). Discovery of a diagnostic biomarker for colon cancer through proteomic profiling of small extracellular vesicles. BMC Cancer, 18(1), 1–11.

    Article  CAS  Google Scholar 

  72. Kenific, C. M., et al. (2010). Autophagy and metastasis: Another double-edged sword. Current Opinion in Cell Biology, 22(2), 241–245.

    Article  CAS  Google Scholar 

  73. Lu, Z., et al. (2008). The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. The Journal of Clinical Investigation, 118(12), 3917–3929.

    CAS  Google Scholar 

  74. Mowers, E. E., et al. (2017). Autophagy in cancer metastasis. Oncogene, 36(12), 1619–1630.

    Article  CAS  Google Scholar 

  75. Su, Z., et al. (2015). Apoptosis, autophagy, necroptosis, and cancer metastasis. Molecular Cancer, 14, 1–14.

    Article  CAS  Google Scholar 

Download references

Acknowledgments

I would like to thank all the students in the lab for their help and the members of the research group for their efforts. I would also like to thank my advisors for their guidance. In addition, thanks for the funding supports of the National Natural Science Fund (Grant No. 82073277 and 82173247), the Science and Technology Project Affiliated to the Education Department of Chongqing (Grant No. KJQN202100404), Natural Science Fund of Chongqing (Grant No. cstc2019jcyj-msxmX0868) and Science and Technology Project of Chongqing Yuzhong District (Grant No. 20200110).

Funding

This work was supported by the National Natural Science Fund (Grant No. 82073277 and 82173247), the Science and Technology Project Affiliated to the Education Department of Chongqing (Grant No. KJQN202100404), Natural Science Fund of Chongqing (Grant No. cstc2019jcyj-msxmX0868) and Science and Technology Project of Chongqing Yuzhong District (Grant No. 20200110).

Author information

Authors and Affiliations

Authors

Contributions

Doudou Liu and Xiaoshuang Li completed the molecular experiment, Bin Zeng and Qiting Zhao completed the animal experiment, Hao Chen was responsible for exosome extraction, Yuhan Zhang and Yuting Chen completed the analysis of sequencing data. Doudou Liu wrote the article, Jianyu Wang and H. Rosie Xing were responsible for the overall design of this study and the revision and improvement of the manuscript.

Corresponding authors

Correspondence to Jianyu Wang or H. Rosie Xing.

Ethics declarations

Competing Interests

The authors declare that they have no competing interests.

Ethical Approval

All animal work was conducted in accordance with an approved protocol and carried out in accordance with the institutional animal welfare guidelines of the Chongqing Medical University. The license number of experimental animal is SYXK2018–0003.

Consent to Participate

All authors agreed to participate in the study of the subject.

Consent for Publication

All authors agree to revise the paper for publication.

Additional information

Publisher’s Note

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

Supplementary Information

Supplementary Figure 1

A. Efficiency of transfection of miR-4535 mimics into MSC exosomes by electroporation, **refers to p < 0.01. B. Expression of miR-4535 in MPC cells was increased after MPC cells co-cultured with MSC exosomes that overexpressed miR-4535, *refers to p < 0.05. C-D. Overexpression of miR-4535 in MSC exosomes promoted the migration (C) and invasion (D) of MPC cells, the bar is 210 μm, **refers to p < 0.01. E. The level of miR-4535 in MSC exosomes was increased when miR-4535 was overexpressed in MSC cells, ***refers to p < 0.001. F. After overexpression of miR-4535 in MSC cells, MSC exosomes were co-cultured with MPC, and the expression of miR-4535 in MPC was also increased, *refers to p < 0.05. G-H. After miR-4535 overexpression in MSC cells, MSC exosomes promoted the migration (G) and invasion (H) of MPC cells, the bar is 210 μm, ***refers to p < 0.001. I. After inhibiting the expression of miR-4535 in MSC cells with the inhibitor, the level of miR-4535 in exosomes was also decreased, ***refers to p < 0.001. (PNG 2269 kb)

High resolution image (TIF 19633 kb)

Supplementary Figure 2

A. Predicted binding site and score of miRNA-4535 and ATG13 in TargetScan. B. Predicted binding sites and mutation sites of miRNA-4535 and ATG13. C. Relative luciferase activities were analyzed in 293 T cells co-transfected with ATG13 3 ′ -UTR (wild type or mutant) reporter plasmid, and microRNA 4535 mimic or NC mimics. (PNG 475 kb)

High resolution image (TIF 9727 kb)

Supplementary Figure 3

A. Assessment of the stemness of MSC cells by the spheroid formation ability of 1000 cells in a 6-well plate. The scale bar is 300 μm. B. Assessment of the stemness of MSC cells by single cell cloning rate in 96-well plates after the expression of miRNA 4535 was inhibited in MSC cells. C. The expression of stemness genes after the expression of miRNA 4535 was inhibited in MSC cells. ***refers to P < 0.001 and **refers to p < 0.01. (PNG 1728 kb)

High resolution image (TIF 68560 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, D., Li, X., Zeng, B. et al. Exosomal microRNA-4535 of Melanoma Stem Cells Promotes Metastasis by Inhibiting Autophagy Pathway. Stem Cell Rev and Rep 19, 155–169 (2023). https://doi.org/10.1007/s12015-022-10358-4

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12015-022-10358-4

Keywords

Navigation