Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Novel lncRNA Gm33149 modulates metastatic heterogeneity in melanoma by regulating the miR-5623-3p/Wnt axis via exosomal transfer

Abstract

The high mortality rate associated with melanoma primarily results from metastasis and recurrence. However, the precise mechanisms driving these processes remain poorly understood. Intercellular communication between cancer cells and non-cancer cells significantly influences the tumor microenvironment and plays a crucial role in metastasis. Therefore, our current study aims to investigate the role and mechanism of long non-coding RNAs (lncRNAs) in regulating the interaction between melanoma cancer stem cells (CSCs) and non-CSCs during the metastatic colonization process. This study has characterized a novel lncRNA called Gm33149. Importantly, we provide evidence for the first time that Gm33149, originating from highly metastatic melanoma stem cells (OL-SD), can be packaged into exosomes and transferred to low-metastatic nonstem cells (OL). Once internalized by OL cells, Gm33149 exerts its function through a competitive endogenous RNA mechanism (ceRNA) involving miR-5623-3p. Specifically, Gm33149 competitively binds to miR-5623-3p, thereby activating the Wnt signaling pathway and promoting the acquisition of a more aggressive metastatic phenotype by OL cells. In summary, our findings suggest that targeting lncRNA Gm33149 within extracellular vesicles could potentially serve as a therapeutic strategy for the treatment of metastatic melanoma.

Schematic representation of the mechanisms underlying the pro-metastatic activity of lncRNA Gm33149 mediated by exosomal transfer. The figure illustrates the key mechanisms involved in the pro-metastatic activity of lncRNA Gm33149 through exosomal transfer. Melanoma stem cells (OLSD) release exosomes containing lncRNA Gm33149. These exosomes are taken up by non-stem melanoma cells (OL), delivering lncRNA Gm33149 to the recipient cells. Within OL cells, lncRNA Gm33149 functions as a competitive endogenous RNA (ceRNA), sequestering miR-5623-3p. This sequestration prevents miR-5623-3p from binding to its target genes, thereby activating the Wnt signaling pathway. The activated Wnt signaling pathway enhances the migration, invasion, and metastatic colonization capabilities of OL cells. The transfer of lncRNA Gm33149 via exosomes contributes to OL cells acquiring “metastatic competency” while promoting their metastatic colonization. These findings underscore the importance of lncRNA Gm33149 in intercellular communication and the metastatic progression of melanoma.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Exosomes secreted by melanoma stem cells (CSCs) can promote the metastasis of no-CSC melanoma cells.
Fig. 2: Prioritization of novel lncRNA Gm33149.
Fig. 3: miR-5623-3p promotes metastatic colonization of non-CSC melanoma cells.
Fig. 4: lncRNA Gm33149 sequesters miR-5623-3p to promote the metastatic colonization of non-CSC melanoma cells.
Fig. 5: The Wnt signaling pathway functions downstream of lncRNA Gm33149 to mediate its pro-metastatic activity.

Similar content being viewed by others

Data availability

The datasets generated during and/or analyzed during the current study are available from the corresponding author upon reasonable request.

References

  1. Ahmed B, Qadir MI, Ghafoor S. Malignant Melanoma: Skin Cancer-Diagnosis, Prevention, and Treatment. Crit Rev Eukaryot Gene Expr. 2020;30:291–7.

    Article  PubMed  Google Scholar 

  2. Kalaora S, Nagler A, Wargo JA, Samuels Y. Mechanisms of immune activation and regulation: lessons from melanoma. Nat Rev Cancer. 2022;22:195–207.

    Article  CAS  PubMed  Google Scholar 

  3. Sanmamed MF, Chen L. Inducible expression of B7-H1 (PD-L1) and its selective role in tumor site immune modulation. Cancer J (Sudbury, Mass). 2014;20:256–61.

    Article  CAS  Google Scholar 

  4. Swoboda A, Soukup R, Eckel O, Kinslechner K, Wingelhofer B, Schörghofer D, et al. STAT3 promotes melanoma metastasis by CEBP-induced repression of the MITF pathway. Oncogene. 2021;40:1091–105.

    Article  CAS  PubMed  Google Scholar 

  5. Botti G, Fratangelo F, Cerrone M, Liguori G, Cantile M, Anniciello AM, et al. COX-2 expression positively correlates with PD-L1 expression in human melanoma cells. J Transl Med. 2017;15:46.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Marzagalli M, Ebelt ND, Manuel ER. Unraveling the crosstalk between melanoma and immune cells in the tumor microenvironment. Semin Cancer Biol. 2019;59:236–50.

    Article  CAS  PubMed  Google Scholar 

  7. Peña-Romero AC, Orenes-Piñero E. Dual Effect of Immune Cells within Tumour Microenvironment: Pro- and Anti-Tumour Effects and Their Triggers. Cancers. 2022;14:1681.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Patton EE, Mueller KL, Adams DJ, Anandasabapathy N, Aplin AE, Bertolotto C, et al. Melanoma models for the next generation of therapies. Cancer cell. 2021;39:610–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, et al. Suppression of Exosomal PD-L1 Induces Systemic Anti-tumor Immunity and Memory. Cell. 2019;177:414–27.e413.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. García-Silva S, Benito-Martín A, Nogués L, Hernández-Barranco A, Mazariegos MS, Santos V, Hergueta-Redondo M, Ximénez-Embún P, et al. Melanoma-derived small extracellular vesicles induce lymphangiogenesis and metastasis through an NGFR-dependent mechanism. Nat cancer. 2021;2:1387–405.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, Zhang G, et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther. 2020;5:8.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Najafi M, Farhood B, Mortezaee K. Cancer stem cells (CSCs) in cancer progression and therapy. J Cell Physiol. 2019;234:8381–95.

    Article  CAS  PubMed  Google Scholar 

  14. Ayob AZ, Ramasamy TS. Cancer stem cells as key drivers of tumour progression. J Biomed Sci. 2018;25:20.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Munro MJ, Wickremesekera SK, Peng L, Tan ST, Itinteang T. Cancer stem cells in colorectal cancer: a review. J Clin Pathol. 2018;71:110–6.

    Article  CAS  PubMed  Google Scholar 

  16. Muraoka D, Seo N, Hayashi T, Tahara Y, Fujii K, Tawara I, et al. Antigen delivery targeted to tumor-associated macrophages overcomes tumor immune resistance, The. J Clin Investig. 2019;129:1278–94.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Eun K, Ham SW, Kim H. Cancer stem cell heterogeneity: origin and new perspectives on CSC targeting. BMB Rep. 2017;50:117–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Liu H, Luo J, Luan S, He C, Li Z. Long non-coding RNAs involved in cancer metabolic reprogramming. Cell Mol Life Sci: CMLS. 2019;76:495–504.

    Article  PubMed  Google Scholar 

  19. Li J, Meng H, Bai Y, Wang K. Regulation of lncRNA and Its Role in Cancer Metastasis. Oncol Res. 2016;23:205–17.

    Article  PubMed  PubMed Central  Google Scholar 

  20. ZHANG ZX. Shi-ping, Research progress on role of Wnt signaling pathway in regulation of tumors[J]. Chin Pharmacol Bull. 2017;33(1):14–17,18.

    Google Scholar 

  21. Gambi G, Mengus G, Davidson G, Demesmaeker E, Cuomo A, Bonaldi T, et al. The lncRNA LENOX Interacts with RAP2C to Regulate Metabolism and Promote Resistance to MAPK Inhibition in Melanoma. Cancer Res. 2022;82:4555–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Li Y, Gao Y, Niu X, Tang M, Li J, Song B, et al. lncRNA BASP1-AS1 interacts with YBX1 to regulate Notch transcription and drives the malignancy of melanoma. Cancer Sci. 2021;112:4526–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Liu Y, Zhuang Y, Fu X, Li C. lncRNA POU3F3 promotes melanoma cell proliferation by downregulating lncRNA MEG3, Discover. Oncology. 2021;12:21.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Duchartre Y, Kim YM, Kahn M. The Wnt signaling pathway in cancer. Crit Rev Oncol/Hematol. 2016;99:141–9.

    Article  PubMed  Google Scholar 

  25. Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene. 2017;36:1461–73.

    Article  CAS  PubMed  Google Scholar 

  26. Zhang Y, Wang X. Targeting the Wnt/β-catenin signaling pathway in cancer. J Hematol Oncol. 2020;13:165.

    Article  PubMed  PubMed Central  Google Scholar 

  27. de Sousa EMF, Vermeulen L. Wnt Signaling in Cancer Stem Cell Biology. Cancers. 2016;8:60.

    Article  Google Scholar 

  28. Zhang Y, Chen Y, Shi L, Li J, Wan W, Li B, et al. Extracellular vesicles microRNA-592 of melanoma stem cells promotes metastasis through activation of MAPK/ERK signaling pathway by targeting PTPN7 in non-stemness melanoma cells. Cell Death Discov. 2022;8:428.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liu D, Li X, Zeng B, Zhao Q, Chen H, Zhang Y, et al. H.R. Xing, Exosomal microRNA-4535 of Melanoma Stem Cells Promotes Metastasis by Inhibiting Autophagy Pathway. Stem cell Rev Rep. 2023;19:155–69.

    Article  CAS  PubMed  Google Scholar 

  30. Li X, Liu D, Chen H, Zeng B, Zhao Q, Zhang Y, et al. H.R. Xing, Melanoma stem cells promote metastasis via exosomal miR-1268a inactivation of autophagy. Biol Res. 2022;55:29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Weiss F, Lauffenburger D, Friedl P. Towards targeting of shared mechanisms of cancer metastasis and therapy resistance. Nat Rev Cancer. 2022;22:157–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang X, Huang J, Chen W, Li G, Li Z, Lei J. The updated role of exosomal proteins in the diagnosis, prognosis, and treatment of cancer. Exp Mol Med. 2022;54:1390–400.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Marte B. Tumour heterogeneity. Nature. 2013;501:327.

    Article  ADS  CAS  PubMed  Google Scholar 

  34. Zhao S, Guan B, Mi Y, Shi D, Wei P, Gu Y, et al. lncRNA MIR17HG promotes colorectal cancer liver metastasis by mediating a glycolysis-associated positive feedback circuit. Oncogene. 2021;40:4709–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Möller A, Lobb RJ. The evolving translational potential of small extracellular vesicles in cancer. Nat Rev Cancer. 2020;20:697–709.

    Article  PubMed  Google Scholar 

  36. Chen F, Chen J, Yang L, Liu J, Zhang X, Zhang Y, Tu Q, et al. Extracellular vesicle-packaged HIF-1α-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat cell Biol. 2019;21:498–510.

    Article  CAS  PubMed  Google Scholar 

  37. Zhang W, Wang Q, Yang Y, Zhou S, Zhang P, Feng T. The role of exosomal lncRNAs in cancer biology and clinical management. Exp Mol Med. 2021;53:1669–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Yu Z, Tang H, Chen S, Xie Y, Shi L, Xia S, et al. Exosomal LOC85009 inhibits docetaxel resistance in lung adenocarcinoma through regulating ATG5-induced autophagy. Drug Resistance Updates Rev Commentaries Antimicrobial Anticancer Chemother. 2023;67:100915.

    CAS  Google Scholar 

  39. Chen H, Zeng B, Li X, Zhao Q, Liu D, Chen Y, et al. High-Metastatic Melanoma Cells Promote the Metastatic Capability of Low-Metastatic Melanoma Cells via Exosomal Transfer of miR-411-5p. Front Oncol. 2022;12:895164.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang CG, Hu YH, Su SL, Zhong D. LncRNA DANCR and miR-320a suppressed osteogenic differentiation in osteoporosis by directly inhibiting the Wnt/β-catenin signaling pathway. Exp Mol Med. 2020;52:1310–25.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Zhang M, Weng W, Zhang Q, Wu Y, Ni S, Tan C, Xu M, Sun H, Liu C, Wei P, Du X. The lncRNA NEAT1 activates Wnt/β-catenin signaling and promotes colorectal cancer progression via interacting with DDX5. J Hematol Oncol. 2018;11:113.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Jang SC, Economides KD, Moniz RJ, Sia CL, Lewis N, McCoy C, et al. ExoSTING, an extracellular vesicle loaded with STING agonists, promotes tumor immune surveillance, Commun. Biol. 2021;4:497.

    CAS  Google Scholar 

  43. Zhang Z, Zhou C, Chang Y, Zhang Z, Hu Y, Zhang F, et al. Long non-coding RNA CASC11 interacts with hnRNP-K and activates the WNT/β-catenin pathway to promote growth and metastasis in colorectal cancer. Cancer Lett. 2016;376:62–73.

    Article  CAS  PubMed  Google Scholar 

  44. Yang XZ, Cheng TT, He QJ, Lei ZY, Chi J, Tang Z, et al. LINC01133 as ceRNA inhibits gastric cancer progression by sponging miR-106a-3p to regulate APC expression and the Wnt/β-catenin pathway. Mol Cancer. 2018;17:126.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Throughout the writing of this dissertation, I have received a great deal of support and assistance. I would first like to thank my supervisor, H. Rosie Xing, whose expertise was invaluable in formulating the research questions and methodology. I would also like to thank my tutor, Jian-Yu Wang, for their valuable guidance throughout my studies. I would particularly like to acknowledge my team members, Yu-Han Zhang, Jie Li, Lei Shi, Jia-Cheng Xie, Xue Han, Yuting Chen, Meng Xiang, and Bo-Wen Li, for their wonderful collaboration and patient support.

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), and Project of Chongqing Natural Science Foundation Innovation and Development Fund (Municipal Education Commission) (Grant No. CSTB2022NS CQ-LZX0023).

Author information

Authors and Affiliations

Authors

Contributions

YC, YH Z and JL conducted the experiments and analyzed the data. YC was responsible for writing the manuscript. LS, JC X, XH, YT C, MX and BW L participated in the conduction of this study. JY W and HR X designed and oversaw the execution of this study, and contributed to the writing of this manuscript.

Corresponding authors

Correspondence to H. Rosie Xing or Jian-Yu Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Consent for publication

The Author confirms: that the work described has not been published before; that it is not under consideration for publication elsewhere; and that its publication has been approved by all authors.

Ethical approval

Animal experiments were approved by the Chongqing Medical University committee for animal experiments. All experiments were performed with relevant guidelines and regulations. Title: Novel lncRNA Gm33149 Modulates Metastatic Heterogeneity in Melanoma by Regulating the miR-5623-3p/Wnt Axis via Exosomal Transfer; the institutional approval committee: Chongqing Medical University committee; approval number: SYXK2018-0003; date: 2022-02-22\2022-06-23.

Additional information

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

Supplementary information

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

Chen, Y., Zhang, YH., Li, J. et al. Novel lncRNA Gm33149 modulates metastatic heterogeneity in melanoma by regulating the miR-5623-3p/Wnt axis via exosomal transfer. Cancer Gene Ther 31, 364–375 (2024). https://doi.org/10.1038/s41417-023-00707-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41417-023-00707-x

This article is cited by

Search

Quick links