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:

Particulate mediators of the bystander effect linked to suicide and interferon-β transgene expression in melanoma cells

Abstract

In the context of comparative oncology, melanoma cells derived from companion animal tumors are good models for optimizing and predicting their in vivo response to therapeutic strategies. Here, we report that human, canine, and feline melanoma cells driven to death by bleomycin, interferon-β gene, or herpes simplex virus thymidine kinase/ganciclovir suicide gene (SG) treatment significantly increased their internal granularity. This fact correlated with the release of a heterogeneous collection of nano- and micro-sized granules as revealed by transmission electron microscopy. While killing lipofected cells, the expressed transgenes and their derived products were incorporated into these granules that were isolated by differential centrifugation. These particulate factors (PFs) were able to transfer, in a dose- and time-dependent manner, appreciable levels of therapeutic genes, related proteins, and drugs. Thus, when recipient cells of SG-carrying PFs were exposed to ganciclovir, this prodrug was efficiently activated, eliminating them. These PFs kept the functionality of their cargo, even after being subjected to adverse conditions, such as the presence of DNase, freezing, or heating. Since our in vitro system did not include any of the immune mechanisms that could provide additional antitumor activity, the chemo-gene treatments amplified by these delivery bags of therapeutic agents offer a great clinical potential.

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: Induction of  intracellular granularity by antitumoral agents.
Fig. 2: Correlations between intracellular granularity and extracellular granules.
Fig. 3: Quantification and cytotoxic effects of the particulate factors released by suicide gene.
Fig. 4: Plasmidic DNAs and their translated proteins carried by particulate factors.
Fig. 5: Direct or particulate factors mediated cytotoxic effects of the treatments.
Fig. 6: Dose, incubation time and heparin effects on particulate factors mediated cytotoxicity.
Fig. 7: Direct or particulate factors mediated cytotoxic effects of treatments.

Similar content being viewed by others

References

  1. Prouteau A, André C. Canine melanomas as models for human melanomas: clinical, histological, and genetic comparison. Genes. 2019;10:501.

  2. Chamel G, Abadie J, Albaric O, Labrut S, Ponce F, Ibisch C. Non-ocular melanomas in cats: a retrospective study of 30 cases. J Feline Med Surg. 2017;19:351–7.

    PubMed  Google Scholar 

  3. Lerner BA, Stewart LA, Horowitz DP, Carvajal RD. Mucosal melanoma: new insights and therapeutic options for a unique and aggressive disease. Oncology. 2017;31:e23–32.

    PubMed  Google Scholar 

  4. Nishida-Aoki N, Gujral TS. Emerging approaches to study cell-cell interactions in tumor microenvironment. Oncotarget. 2019;10:785–97.

    PubMed  PubMed Central  Google Scholar 

  5. Zhang HG, Grizzle WE. Exosomes: a novel pathway of local and distant intercellular communication that facilitates the growth and metastasis of neoplastic lesions. Am J Pathol. 2014;184:28–41.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. 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.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Demirsoy S, Martin S, Maes H, Agostinis P. Adapt, recycle, and move on: proteostasis and trafficking mechanisms in melanoma. Front Oncol. 2016;6:240.

    PubMed  PubMed Central  Google Scholar 

  8. Lv MM, Zhu XY, Chen WX, Zhong SL, Hu Q, Ma TF, et al. Exosomes mediate drug resistance transfer in MCF-7 breast cancer cells and a probable mechanism is delivery of P-glycoprotein. Tumour Biol. 2014;35:10773–9.

    CAS  PubMed  Google Scholar 

  9. Samuel P, Mulcahy LA, Furlong F, McCarthy HO, Brooks SA, Fabbri M, et al. Cisplatin induces the release of extracellular vesicles from ovarian cancer cells that can induce invasiveness and drug resistance in bystander cells. Philos Trans R Soc Lond B Biol Sci. 2018;373:20170065.

  10. Emam SE, Ando H, Abu Lila AS, Kobayashi S, Shimizu T, Okuhira K, et al. Doxorubicin expands in vivo secretion of circulating exosome in mice. Biol Pharm Bull. 2018;41:1078–83.

    CAS  PubMed  Google Scholar 

  11. Martinez VG, O’Neill S, Salimu J, Breslin S, Clayton A, Crown J. et al. Resistance to HER2-targeted anti-cancer drugs is associated with immune evasion in cancer cells and their derived extracellular vesicles. Oncoimmunology.2017;6:e1362530

    PubMed  PubMed Central  Google Scholar 

  12. Ciravolo V, Huber V, Ghedini GC, Venturelli E, Bianchi F, Campiglio M, et al. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J Cell Physiol. 2012;227:658–67.

    CAS  PubMed  Google Scholar 

  13. Finocchiaro LM, Fiszman GL, Karara AL, Glikin GC. Suicide gene and cytokines combined nonviral gene therapy for spontaneous canine melanoma. Cancer Gene Ther. 2008;15:165–72.

    CAS  PubMed  Google Scholar 

  14. Finocchiaro LM, Glikin GC. Cytokine-enhanced vaccine and suicide gene therapy as surgery adjuvant treatments for spontaneous canine melanoma: 9 years of follow-up. Cancer Gene Ther. 2012;19:852–61.

    CAS  PubMed  Google Scholar 

  15. Finocchiaro LM, Fondello C, Gil-Cardeza ML, Rossi Ú, Villaverde MS, Riveros MD, et al. Cytokine-enhanced vaccine and interferon-β plus suicide gene therapy as surgery adjuvant treatments for spontaneous canine melanoma. Hum Gene Ther. 2015;26:367–76.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Gil-Cardeza ML, Villaverde MS, Fiszman GL, Altamirano NA, Cwirenbaum RA, Glikin GC, et al. Suicide gene therapy on spontaneous canine melanoma: correlations between in vivo tumors and their derived multicell spheroids in vitro. Gene Ther. 2010;17:26–36.

    CAS  PubMed  Google Scholar 

  17. Agnetti L, Fondello C, Villaverde MS, Glikin GC, Finocchiaro LME. Therapeutic potential of bleomycin plus suicide or interferon-β gene transfer combination for spontaneous feline and canine melanoma. Oncoscience. 2017;4:199–214.

    PubMed  PubMed Central  Google Scholar 

  18. Fondello C, Agnetti L, Villaverde MS, Simian M, Glikin GC, Finocchiaro LME. The combination of bleomycin with suicide or interferon-β gene transfer is able to efficiently eliminate human melanoma tumor initiating cells. Biomed Pharmacother. 2016;83:290–301.

    CAS  PubMed  Google Scholar 

  19. Finocchiaro LME, Agnetti L, Fondello C, Glikin GC. Combination of cytokine-enhanced vaccine and chemo-gene therapy as surgery adjuvant treatments for spontaneous canine melanoma. Gene Ther. 2019;26:418–31.

    CAS  PubMed  Google Scholar 

  20. Rossi Ú, Gil-Cardeza ML, Villaverde MS, Finocchiaro LM, Glikin GC. Interferon-β gene transfer induces a strong cytotoxic bystander effect on melanoma cells. Biomed Pharmacother. 2015;72:44–51.

    CAS  PubMed  Google Scholar 

  21. Wu K, Xing F, Wu SY, Watabe K. Extracellular vesicles as emerging targets in cancer: recent development from bench to bedside. Biochim Biophys Acta Rev Cancer. 2017;1868:538–63.

    CAS  PubMed  Google Scholar 

  22. Turturici G, Tinnirello R, Sconzo G, Geraci F. Extracellular membrane vesicles as a mechanism of cell-to-cell communication: advantages and disadvantages. Am J Physiol Cell Physiol. 2014;306:C621–33.

    CAS  PubMed  Google Scholar 

  23. Minciacchi VR, Freeman MR, Di Vizio D. Extracellular vesicles in cancer: exosomes, microvesicles and the emerging role of large oncosomes. Semin Cell Dev Biol. 2015;40:41–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Maas SLN, Breakefield XO, Weaver AM. Extracellular vesicles: unique intercellular delivery vehicles . Trends Cell Biol. 2017;27:172–88.

    CAS  PubMed  Google Scholar 

  25. Jella KK, Rani S, O’Driscoll L, McClean B, Byrne HJ, Lyng FM. Exosomes are involved in mediating radiation induced bystander signaling in human keratinocyte cells. Radiat Res. 2014;181:138–45.

    CAS  PubMed  Google Scholar 

  26. Hirt B. Selective extraction of polyoma DNA from infected mouse cell cultures. J Mol Biol. 1967;26:365–9.

    CAS  PubMed  Google Scholar 

  27. Haynes MK, Strouse JJ, Waller A, Leitao A, Curpan RF, Bologa C, et al. Detection of intracellular granularity induction in prostate cancer cell lines by small molecules using the HyperCyt high-throughput flow cytometry system. J Biomol Screen. 2009;14:596–609.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Gil-Cardeza ML, Rossi Ú, Villaverde MS, Glikin GC, Finocchiaro LM. Cationic lipid:DNA complexes allow bleomycin uptake by melanoma cells. Biomed Pharmacother. 2013;67:269–75.

    CAS  PubMed  Google Scholar 

  29. Chinnadurai M, Chidambaram S, Ganesan V, Baraneedharan U, Sundaram L, Paul SF, et al. Bleomycin, neocarzinostatin and ionising radiation-induced bystander effects in normal diploid human lung fibroblasts, bone marrow mesenchymal stem cells, lung adenocarcinoma cells and peripheral blood lymphocytes. Int J Radiat Biol. 2011;87:673–82.

    CAS  PubMed  Google Scholar 

  30. Savu D, Petcu I, Temelie M, Mustaciosu C, Moisoi N. Compartmental stress responses correlate with cell survival in bystander effects induced by the DNA damage agent, bleomycin. Mutat Res. 2015;771:13–20.

    CAS  PubMed  Google Scholar 

  31. Atai NA, Balaj L, van Veen H, Breakefield XO, Jarzyna PA, Van Noorden CJ, et al. Heparin blocks transfer of extracellular vesicles between donor and recipient cells. J Neurooncol. 2013;115:343–51.

    CAS  PubMed  Google Scholar 

  32. Christianson HC, Svensson KJ, van Kuppevelt TH, Li JP, Belting M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Proc Natl Acad Sci USA. 2013;110:17380–5.

    CAS  PubMed  Google Scholar 

  33. Moon H, Donahue LR, Choi E, Scumpia PO, Lowry WE, Grenier JK, et al. Mela-nocyte stem cell activation and translocation initiate cutaneous melanoma in response to UV exposure. Cell Stem Cell. 2017;21:665–78.e6.

    CAS  PubMed  Google Scholar 

  34. Santiago-Walker A, Li L, Haass NK, Herlyn M. Melanocytes: from morphology to application. Skin Pharmacol Physiol. 2009;22:114–21.

    CAS  PubMed  Google Scholar 

  35. Cichorek M, Wachulska M, Stasiewicz A, Tymińska A. Skin melanocytes: biology and development. Postepy Dermatol Alergol. 2013;30:30–41.

  36. Karjoo Z, Chen X, Hatefi A. Progress and problems with the use of suicide genes for targeted cancer therapy. Adv Drug Deliv Rev. 2016;99:113–28.

    CAS  PubMed  Google Scholar 

  37. Krasagakis K, Garbe C, Krüger S, Orfanos CE. Effects of interferons on cultured human melanocytes in vitro: interferon-beta but not-alpha or –gamma inhibit proliferation and all interferons significantly modulate the cell phenotype. J Investig Dermatol. 1991;97:364–72.

    CAS  PubMed  Google Scholar 

  38. Yang J, Liu TJ, Lu Y. Effects of bicistronic lentiviral vector-mediated herpes simplex virus thymidine kinase/ganciclovir system on human lens epithelial cells. Curr Eye Res. 2007;32:33–42.

    PubMed  Google Scholar 

  39. Danesi CC, Dihl RR, Bellagamba BC, de Andrade HH, Cunha KS, Guimarães NN, et al. Genotoxicity testing of combined treatment with cisplatin, bleomycin, and 5-fluorouracil in somatic cells of Drosophila melanogaster. Mutat Res. 2012;747:228–33.

    CAS  PubMed  Google Scholar 

  40. Altanerova U, Jakubechova J, Benejova K, Priscakova P, Pesta M, Pitule P, et al. Prodrug suicide gene therapy for cancer targeted intracellular by mesenchymal stem cell exosomes. Int J Cancer. 2019;144:897–908.

    PubMed  Google Scholar 

  41. Erkan EP, Saydam N, Chen CC, Saydam O. Extracellular vesicles as carriers of suicide mRNA and/or protein in cancer therapy. Methods Mol Biol. 2019;1895:87–96.

    CAS  PubMed  Google Scholar 

  42. Mizrak A, Bolukbasi MF, Ozdener GB, Brenner GJ, Madlener S, Erkan EP, et al. Genetically engineered microvesicles carrying suicide mRNA/protein inhibit schwannoma tumor growth. Mol Ther. 2013;21:101–8.

    CAS  PubMed  Google Scholar 

  43. Erkan EP, Senfter D, Madlener S, Jungwirth G, Ströbel T, Saydam N, et al. Extracellular vesicle-mediated suicide mRNA/protein delivery inhibits glioblastoma tumor growth in vivo. Cancer Gene Ther. 2017;24:38–44.

    CAS  PubMed  Google Scholar 

  44. Villagrasa A, Álvarez PJ, Osuna A, Garrido JM, Aránega A, Rodríguez-Serrano F. Exosomes derived from breast cancer cells, small Trojan horses? J Mammary Gland Biol Neoplasia. 2014;19:303–13.

    PubMed  Google Scholar 

  45. Hsu DH, Paz P, Villaflor G, Rivas A, Mehta-Damani A, Angevin E, et al. Exosomes as a tumor vaccine: enhancing potency through direct loading of antigenic peptides. J Immunother. 2003;26:440–50.

    CAS  PubMed  Google Scholar 

  46. Zhang H, Huang B. Tumor cell-derived microparticles: a new form of cancer vaccine. Oncoimmunology. 2015;4:e1017704.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Juan M. Cardini and Graciela B. Zenobi for technical assistance. This study was partially supported by grants from ANPCYT/FONCYT (PICT2014-1652) and CONICET (PIP 11220150100885). GCG and LMEF are investigators, and LA, CF, and MFA fellows of the Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET, Argentina).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Liliana M. E. Finocchiaro.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Agnetti, L., Fondello, C., Arbe, M. et al. Particulate mediators of the bystander effect linked to suicide and interferon-β transgene expression in melanoma cells. Gene Ther 28, 38–55 (2021). https://doi.org/10.1038/s41434-020-0136-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41434-020-0136-x

Search

Quick links