3D Cell Cultures as Prospective Models to Study Extracellular Vesicles in Cancer
Abstract
:Simple Summary
Abstract
1. Introduction
2. The 3D Cultures as a Physiological Model of Tumoral Cells
3. Extracellular Vesicles in Cancer Research
4. Production of EVs in 3D Cultures
5. Modelling the Antitumoral Effect of EVs in 3D Cultures
6. EVs-Mediated Crosstalk between the Tumor and Cellular Matrix
6.1. Tumoral Cells Modify Surroundings Cells through EVs
6.2. Tumoral EVs Modify the Tumoral Cells
6.3. Effect of EVs Released by the Cellular Component of the Matrix over Tumor Cells
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Meehan, K.; Vella, L.J. The contribution of tumour-derived exosomes to the hallmarks of cancer. Crit. Rev. Clin. Lab. Sci. 2016, 53, 121–131. [Google Scholar] [CrossRef]
- Lv, D.; Hu, Z.; Lu, L.; Lu, H.; Xu, X. Three-dimensional cell culture: A powerful tool in tumor research and drug discovery. Oncol. Lett. 2017, 14, 6999–7010. [Google Scholar] [CrossRef] [Green Version]
- Weigelt, B.; Ghajar, C.M.; Bissell, M.J. The need for complex 3d culture models to unravel novel pathways and identify accurate biomarkers in breast cancer. Adv. Drug Deliv. Rev. 2014, 69–70, 42–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Katsuda, T.; Kosaka, N.; Ochiya, T. The roles of extracellular vesicles in cancer biology: Toward the development of novel cancer biomarkers. Proteomics 2014, 14, 412–425. [Google Scholar] [CrossRef] [PubMed]
- Ruivo, C.F.; Adem, B.; Silva, M.; Melo, S.A. The biology of cancer exosomes: Insights and new perspectives. Cancer Res. 2017, 77, 6480–6488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tai, Y.L.; Lin, C.J.; Li, T.K.; Shen, T.L.; Hsieh, J.T.; Chen, B.P.C. The role of extracellular vesicles in prostate cancer with clinical applications. Endocr. Relat. Cancer 2020, 27, R133–R144. [Google Scholar] [CrossRef]
- Birgersdotter, A.; Sandberg, R.; Ernberg, I. Gene expression perturbation in vitro--a growing case for three-dimensional (3d) culture systems. Semin. Cancer Biol. 2005, 15, 405–412. [Google Scholar] [CrossRef]
- Kapalczynska, M.; Kolenda, T.; Przybyla, W.; Zajaczkowska, M.; Teresiak, A.; Filas, V.; Ibbs, M.; Blizniak, R.; Luczewski, L.; Lamperska, K. 2d and 3d cell cultures—A comparison of different types of cancer cell cultures. Arch. Med. Sci. 2018, 14, 910–919. [Google Scholar] [CrossRef]
- Fiorini, E.; Veghini, L.; Corbo, V. Modeling cell communication in cancer with organoids: Making the complex simple. Front. Cell Dev. Biol. 2020, 8, 166. [Google Scholar] [CrossRef] [Green Version]
- Ravi, M.; Paramesh, V.; Kaviya, S.R.; Anuradha, E.; Solomon, F.D. 3D cell culture systems: Advantages and applications. J. Cell Physiol. 2015, 230, 16–26. [Google Scholar] [CrossRef]
- Place, E.S.; George, J.H.; Williams, C.K.; Stevens, M.M. Synthetic polymer scaffolds for tissue engineering. Chem. Soc. Rev. 2009, 38, 1139–1151. [Google Scholar] [CrossRef] [PubMed]
- Mehta, G.; Hsiao, A.Y.; Ingram, M.; Luker, G.D.; Takayama, S. Opportunities and challenges for use of tumor spheroids as models to test drug delivery and efficacy. J. Control Release 2012, 164, 192–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ryan, S.L.; Baird, A.M.; Vaz, G.; Urquhart, A.J.; Senge, M.; Richard, D.J.; O’Byrne, K.J.; Davies, A.M. Drug discovery approaches utilizing three-dimensional cell culture. Assay Drug Dev. Technol. 2016, 14, 19–28. [Google Scholar] [CrossRef] [PubMed]
- Joshi, P.; Lee, M.Y. High content imaging (hci) on miniaturized three-dimensional (3d) cell cultures. Biosensors (Basel) 2015, 5, 768–790. [Google Scholar] [CrossRef] [Green Version]
- Salo, T.; Dourado, M.R.; Sundquist, E.; Apu, E.H.; Alahuhta, I.; Tuomainen, K.; Vasara, J.; Al-Samadi, A. Organotypic three-dimensional assays based on human leiomyoma-derived matrices. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2018, 373. [Google Scholar] [CrossRef]
- Eiraku, M.; Sasai, Y. Self-formation of layered neural structures in three-dimensional culture of es cells. Curr. Opin. Neurobiol. 2012, 22, 768–777. [Google Scholar] [CrossRef]
- Fujii, M.; Shimokawa, M.; Date, S.; Takano, A.; Matano, M.; Nanki, K.; Ohta, Y.; Toshimitsu, K.; Nakazato, Y.; Kawasaki, K.; et al. A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis. Cell Stem Cell 2016, 18, 827–838. [Google Scholar] [CrossRef] [Green Version]
- Neal, J.T.; Li, X.; Zhu, J.; Giangarra, V.; Grzeskowiak, C.L.; Ju, J.; Liu, I.H.; Chiou, S.H.; Salahudeen, A.A.; Smith, A.R.; et al. Organoid modeling of the tumor immune microenvironment. Cell 2018, 175, 1972–1988.e1916. [Google Scholar] [CrossRef] [Green Version]
- Sato, T.; Stange, D.E.; Ferrante, M.; Vries, R.G.; Van Es, J.H.; Van den Brink, S.; Van Houdt, W.J.; Pronk, A.; Van Gorp, J.; Siersema, P.D.; et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and barrett’s epithelium. Gastroenterology 2011, 141, 1762–1772. [Google Scholar] [CrossRef]
- Broutier, L.; Mastrogiovanni, G.; Verstegen, M.M.; Francies, H.E.; Gavarro, L.M.; Bradshaw, C.R.; Allen, G.E.; Arnes-Benito, R.; Sidorova, O.; Gaspersz, M.P.; et al. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat. Med. 2017, 23, 1424–1435. [Google Scholar] [CrossRef]
- Corro, C.; Novellasdemunt, L.; Li, V.S.W. A brief history of organoids. Am. J. Physiol. Cell Physiol. 2020, 319, C151–C165. [Google Scholar] [CrossRef] [PubMed]
- Datta, P.; Dey, M.; Ataie, Z.; Unutmaz, D.; Ozbolat, I.T. 3d bioprinting for reconstituting the cancer microenvironment. NPJ Precis Oncol. 2020, 4, 18. [Google Scholar] [CrossRef] [PubMed]
- Burdett, E.; Kasper, F.K.; Mikos, A.G.; Ludwig, J.A. Engineering tumors: A tissue engineering perspective in cancer biology. Tissue Eng. Part B Rev. 2010, 16, 351–359. [Google Scholar] [CrossRef] [PubMed]
- Haycock, J.W. 3d cell culture: A review of current approaches and techniques. Methods Mol. Biol. 2011, 695, 1–15. [Google Scholar] [CrossRef]
- Carletti, E.; Motta, A.; Migliaresi, C. Scaffolds for tissue engineering and 3d cell culture. Methods Mol. Biol. 2011, 695, 17–39. [Google Scholar] [CrossRef]
- Yan, X.; Zhou, L.; Wu, Z.; Wang, X.; Chen, X.; Yang, F.; Guo, Y.; Wu, M.; Chen, Y.; Li, W.; et al. High throughput scaffold-based 3d micro-tumor array for efficient drug screening and chemosensitivity testing. Biomaterials 2019, 198, 167–179. [Google Scholar] [CrossRef]
- Patra, C.; Talukdar, S.; Novoyatleva, T.; Velagala, S.R.; Muhlfeld, C.; Kundu, B.; Kundu, S.C.; Engel, F.B. Silk protein fibroin from antheraea mylitta for cardiac tissue engineering. Biomaterials 2012, 33, 2673–2680. [Google Scholar] [CrossRef] [Green Version]
- Pedron, S.; Becka, E.; Harley, B.A. Regulation of glioma cell phenotype in 3d matrices by hyaluronic acid. Biomaterials 2013, 34, 7408–7417. [Google Scholar] [CrossRef]
- Lv, D.; Yu, S.C.; Ping, Y.F.; Wu, H.; Zhao, X.; Zhang, H.; Cui, Y.; Chen, B.; Zhang, X.; Dai, J.; et al. A three-dimensional collagen scaffold cell culture system for screening anti-glioma therapeutics. Oncotarget 2016, 7, 56904–56914. [Google Scholar] [CrossRef] [Green Version]
- Dunne, L.W.; Huang, Z.; Meng, W.; Fan, X.; Zhang, N.; Zhang, Q.; An, Z. Human decellularized adipose tissue scaffold as a model for breast cancer cell growth and drug treatments. Biomaterials 2014, 35, 4940–4949. [Google Scholar] [CrossRef]
- Stratmann, A.T.; Fecher, D.; Wangorsch, G.; Gottlich, C.; Walles, T.; Walles, H.; Dandekar, T.; Dandekar, G.; Nietzer, S.L. Establishment of a human 3d lung cancer model based on a biological tissue matrix combined with a boolean in silico model. Mol. Oncol. 2014, 8, 351–365. [Google Scholar] [CrossRef] [PubMed]
- Gilkes, D.M.; Semenza, G.L.; Wirtz, D. Hypoxia and the extracellular matrix: Drivers of tumour metastasis. Nat. Rev. Cancer 2014, 14, 430–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brancato, V.; Garziano, A.; Gioiella, F.; Urciuolo, F.; Imparato, G.; Panzetta, V.; Fusco, S.; Netti, P.A. 3d is not enough: Building up a cell instructive microenvironment for tumoral stroma microtissues. Acta Biomater. 2017, 47, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Zhu, L.; Fan, X.; Wang, B.; Liu, L.; Yan, X.; Zhou, L.; Zeng, Y.; Poznansky, M.C.; Wang, L.; Chen, H.; et al. Biomechanically primed liver microtumor array as a high-throughput mechanopharmacological screening platform for stroma-reprogrammed combinatorial therapy. Biomaterials 2017, 124, 12–24. [Google Scholar] [CrossRef] [PubMed]
- Kleinman, H.K.; Martin, G.R. Matrigel: Basement membrane matrix with biological activity. Semin. Cancer Biol. 2005, 15, 378–386. [Google Scholar] [CrossRef] [PubMed]
- Cukierman, E.; Pankov, R.; Yamada, K.M. Cell interactions with three-dimensional matrices. Curr. Opin. Cell Biol. 2002, 14, 633–639. [Google Scholar] [CrossRef]
- Xu, X.; Sabanayagam, C.R.; Harrington, D.A.; Farach-Carson, M.C.; Jia, X. A hydrogel-based tumor model for the evaluation of nanoparticle-based cancer therapeutics. Biomaterials 2014, 35, 3319–3330. [Google Scholar] [CrossRef] [Green Version]
- Mahoney, C.M.; Kelmindi-Doko, A.; Snowden, M.J.; Peter Rubin, J.; Marra, K.G. Adipose derived delivery vehicle for encapsulated adipogenic factors. Acta Biomater. 2017, 58, 26–33. [Google Scholar] [CrossRef]
- Poupot, M.; Fournie, J.J. Spontaneous membrane transfer through homotypic synapses between lymphoma cells. J. Immunol. 2003, 171, 2517–2523. [Google Scholar] [CrossRef] [Green Version]
- Savina, A.; Vidal, M.; Colombo, M.I. The exosome pathway in k562 cells is regulated by rab11. J. Cell Sci. 2002, 115, 2505–2515. [Google Scholar]
- Maia, J.; Caja, S.; Strano Moraes, M.C.; Couto, N.; Costa-Silva, B. Exosome-based cell-cell communication in the tumor microenvironment. Front. Cell Dev. Biol. 2018, 6, 18. [Google Scholar] [CrossRef] [PubMed]
- Shao, Y.; Chen, T.; Zheng, X.; Yang, S.; Xu, K.; Chen, X.; Xu, F.; Wang, L.; Shen, Y.; Wang, T.; et al. Colorectal cancer-derived small extracellular vesicles establish an inflammatory premetastatic niche in liver metastasis. Carcinogenesis 2018, 39, 1368–1379. [Google Scholar] [CrossRef] [PubMed]
- Fong, M.Y.; Zhou, W.; Liu, L.; Alontaga, A.Y.; Chandra, M.; Ashby, J.; Chow, A.; O’Connor, S.T.; Li, S.; Chin, A.R.; et al. Breast-cancer-secreted mir-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 2015, 17, 183–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robado de Lope, L.; Alcibar, O.L.; Amor Lopez, A.; Hergueta-Redondo, M.; Peinado, H. Tumour-adipose tissue crosstalk: Fuelling tumour metastasis by extracellular vesicles. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2018, 373. [Google Scholar] [CrossRef] [PubMed]
- Kosaka, N.; Yoshioka, Y.; Fujita, Y.; Ochiya, T. Versatile roles of extracellular vesicles in cancer. J. Clin. Invest. 2016, 126, 1163–1172. [Google Scholar] [CrossRef] [Green Version]
- Tai, Y.L.; Chen, K.C.; Hsieh, J.T.; Shen, T.L. Exosomes in cancer development and clinical applications. Cancer Sci. 2018, 109, 2364–2374. [Google Scholar] [CrossRef] [Green Version]
- Ferguson, S.; Weissleder, R. Modeling ev kinetics for use in early cancer detection. Adv. Biosyst. 2020, 4, e1900305. [Google Scholar] [CrossRef]
- Takahashi, K.; Ota, Y.; Kogure, T.; Suzuki, Y.; Iwamoto, H.; Yamakita, K.; Kitano, Y.; Fujii, S.; Haneda, M.; Patel, T.; et al. Circulating extracellular vesicle-encapsulated hulc is a potential biomarker for human pancreatic cancer. Cancer Sci. 2020, 111, 98–111. [Google Scholar] [CrossRef] [Green Version]
- Ma, C.; Jiang, F.; Ma, Y.; Wang, J.; Li, H.; Zhang, J. Isolation and detection technologies of extracellular vesicles and application on cancer diagnostic. Dose Response 2019, 17, 1559325819891004. [Google Scholar] [CrossRef] [Green Version]
- Herrero, C.; de la Fuente, A.; Casas-Arozamena, C.; Sebastian, V.; Prieto, M.; Arruebo, M.; Abalo, A.; Colas, E.; Moreno-Bueno, G.; Gil-Moreno, A.; et al. Extracellular vesicles-based biomarkers represent a promising liquid biopsy in endometrial cancer. Cancers (Basel) 2019, 11, 2000. [Google Scholar] [CrossRef] [Green Version]
- Pang, B.; Zhu, Y.; Ni, J.; Thompson, J.; Malouf, D.; Bucci, J.; Graham, P.; Li, Y. Extracellular vesicles: The next generation of biomarkers for liquid biopsy-based prostate cancer diagnosis. Theranostics 2020, 10, 2309–2326. [Google Scholar] [CrossRef] [PubMed]
- Joncas, F.H.; Lucien, F.; Rouleau, M.; Morin, F.; Leong, H.S.; Pouliot, F.; Fradet, Y.; Gilbert, C.; Toren, P. Plasma extracellular vesicles as phenotypic biomarkers in prostate cancer patients. Prostate 2019, 79, 1767–1776. [Google Scholar] [CrossRef] [PubMed]
- Yekula, A.; Muralidharan, K.; Kang, K.M.; Wang, L.; Balaj, L.; Carter, B.S. From laboratory to clinic: Translation of extracellular vesicle based cancer biomarkers. Methods 2020, 177, 58–66. [Google Scholar] [CrossRef] [PubMed]
- Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of sirna to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
- Tian, Y.; Li, S.; Song, J.; Ji, T.; Zhu, M.; Anderson, G.J.; Wei, J.; Nie, G. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 2014, 35, 2383–2390. [Google Scholar] [CrossRef]
- Bellavia, D.; Raimondo, S.; Calabrese, G.; Forte, S.; Cristaldi, M.; Patinella, A.; Memeo, L.; Manno, M.; Raccosta, S.; Diana, P.; et al. Interleukin 3- receptor targeted exosomes inhibit in vitro and in vivo chronic myelogenous leukemia cell growth. Theranostics 2017, 7, 1333–1345. [Google Scholar] [CrossRef]
- Shao, J.; Zaro, J.; Shen, Y. Advances in exosome-based drug delivery and tumor targeting: From tissue distribution to intracellular fate. Int. J. Nanomed. 2020, 15, 9355–9371. [Google Scholar] [CrossRef]
- Yang, J.; Zhang, Z.; Zhang, Y.; Ni, X.; Zhang, G.; Cui, X.; Liu, M.; Xu, C.; Zhang, Q.; Zhu, H.; et al. Zip4 promotes muscle wasting and cachexia in mice with orthotopic pancreatic tumors by stimulating rab27b-regulated release of extracellular vesicles from cancer cells. Gastroenterology 2019, 156, 722–734.e726. [Google Scholar] [CrossRef] [Green Version]
- Hwang, W.L.; Lan, H.Y.; Cheng, W.C.; Huang, S.C.; Yang, M.H. Tumor stem-like cell-derived exosomal rnas prime neutrophils for facilitating tumorigenesis of colon cancer. J. Hematol. Oncol. 2019, 12, 10. [Google Scholar] [CrossRef] [Green Version]
- Szvicsek, Z.; Oszvald, A.; Szabo, L.; Sandor, G.O.; Kelemen, A.; Soos, A.A.; Paloczi, K.; Harsanyi, L.; Tolgyes, T.; Dede, K.; et al. Extracellular vesicle release from intestinal organoids is modulated by apc mutation and other colorectal cancer progression factors. Cell Mol. Life Sci. 2019, 76, 2463–2476. [Google Scholar] [CrossRef] [Green Version]
- Namba, Y.; Sogawa, C.; Okusha, Y.; Kawai, H.; Itagaki, M.; Ono, K.; Murakami, J.; Aoyama, E.; Ohyama, K.; Asaumi, J.I.; et al. Depletion of lipid efflux pump abcg1 triggers the intracellular accumulation of extracellular vesicles and reduces aggregation and tumorigenesis of metastatic cancer cells. Front. Oncol. 2018, 8, 376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Franchi, M.; Piperigkou, Z.; Karamanos, K.A.; Franchi, L.; Masola, V. Extracellular matrix-mediated breast cancer cells morphological alterations, invasiveness, and microvesicles/exosomes release. Cells 2020, 9, 2031. [Google Scholar] [CrossRef] [PubMed]
- Eguchi, T.; Sogawa, C.; Okusha, Y.; Uchibe, K.; Iinuma, R.; Ono, K.; Nakano, K.; Murakami, J.; Itoh, M.; Arai, K.; et al. Organoids with cancer stem cell-like properties secrete exosomes and hsp90 in a 3d nanoenvironment. PLoS ONE 2018, 13, e0191109. [Google Scholar] [CrossRef] [Green Version]
- Thippabhotla, S.; Zhong, C.; He, M. 3d cell culture stimulates the secretion of in vivo like extracellular vesicles. Sci. Rep. 2019, 9, 13012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tauro, B.J.; Greening, D.W.; Mathias, R.A.; Mathivanan, S.; Ji, H.; Simpson, R.J. Two distinct populations of exosomes are released from lim1863 colon carcinoma cell-derived organoids. Mol. Cell Proteom. 2013, 12, 587–598. [Google Scholar] [CrossRef] [Green Version]
- Huang, L.; Bockorny, B.; Paul, I.; Akshinthala, D.; Frappart, P.O.; Gandarilla, O.; Bose, A.; Sanchez-Gonzalez, V.; Rouse, E.E.; Lehoux, S.D.; et al. Pdx-derived organoids model in vivo drug response and secrete biomarkers. JCI Insight 2020, 5. [Google Scholar] [CrossRef]
- Guerreiro, E.M.; Vestad, B.; Steffensen, L.A.; Aass, H.C.D.; Saeed, M.; Ovstebo, R.; Costea, D.E.; Galtung, H.K.; Soland, T.M. Efficient extracellular vesicle isolation by combining cell media modifications, ultrafiltration, and size-exclusion chromatography. PLoS ONE 2018, 13, e0204276. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Z.; McGill, J.; Gamero-Kubota, P.; He, M. Microfluidic on-demand engineering of exosomes towards cancer immunotherapy. Lab. Chip 2019, 19, 1877–1886. [Google Scholar] [CrossRef]
- Patel, D.B.; Luthers, C.R.; Lerman, M.J.; Fisher, J.P.; Jay, S.M. Enhanced extracellular vesicle production and ethanol-mediated vascularization bioactivity via a 3d-printed scaffold-perfusion bioreactor system. Acta Biomater. 2019, 95, 236–244. [Google Scholar] [CrossRef]
- Ramamoorthy, P.; Thomas, S.M.; Kaushik, G.; Subramaniam, D.; Chastain, K.M.; Dhar, A.; Tawfik, O.; Kasi, A.; Sun, W.; Ramalingam, S.; et al. Metastatic tumor-in-a-dish, a novel multicellular organoid to study lung colonization and predict therapeutic response. Cancer Res. 2019, 79, 1681–1695. [Google Scholar] [CrossRef] [Green Version]
- Murgoci, A.N.; Cizkova, D.; Majerova, P.; Petrovova, E.; Medvecky, L.; Fournier, I.; Salzet, M. Brain-cortex microglia-derived exosomes: Nanoparticles for glioma therapy. Chemphyschem 2018, 19, 1205–1214. [Google Scholar] [CrossRef] [PubMed]
- Yeon, J.H.; Jeong, H.E.; Seo, H.; Cho, S.; Kim, K.; Na, D.; Chung, S.; Park, J.; Choi, N.; Kang, J.Y. Cancer-derived exosomes trigger endothelial to mesenchymal transition followed by the induction of cancer-associated fibroblasts. Acta Biomater. 2018, 76, 146–153. [Google Scholar] [CrossRef]
- Jeong, G.S.; Han, S.; Shin, Y.; Kwon, G.H.; Kamm, R.D.; Lee, S.H.; Chung, S. Sprouting angiogenesis under a chemical gradient regulated by interactions with an endothelial monolayer in a microfluidic platform. Anal. Chem. 2011, 83, 8454–8459. [Google Scholar] [CrossRef]
- Jeong, K.; Yu, Y.J.; You, J.Y.; Rhee, W.J.; Kim, J.A. Exosome-mediated microrna-497 delivery for anti-cancer therapy in a microfluidic 3d lung cancer model. Lab. Chip 2020, 20, 548–557. [Google Scholar] [CrossRef] [PubMed]
- Sancho-Albero, M.; Medel-Martínez, A.; Martín-Duque, P. Use of exosomes as vectors to carry advanced therapies. RSC Advances 2020, 10, 23975–23987. [Google Scholar] [CrossRef]
- Villa, F.; Quarto, R.; Tasso, R. Extracellular vesicles as natural, safe and efficient drug delivery systems. Pharmaceutics 2019, 11, 557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nooshabadi, V.T.; Khanmohammadi, M.; Shafei, S.; Banafshe, H.R.; Malekshahi, Z.V.; Ebrahimi-Barough, S.; Ai, J. Impact of atorvastatin loaded exosome as an anti-glioblastoma carrier to induce apoptosis of u87 cancer cells in 3d culture model. Biochem. Biophys. Rep. 2020, 23, 100792. [Google Scholar] [CrossRef] [PubMed]
- Millard, M.; Yakavets, I.; Piffoux, M.; Brun, A.; Gazeau, F.; Guigner, J.M.; Jasniewski, J.; Lassalle, H.P.; Wilhelm, C.; Bezdetnaya, L. Mthpc-loaded extracellular vesicles outperform liposomal and free mthpc formulations by an increased stability, drug delivery efficiency and cytotoxic effect in tridimensional model of tumors. Drug Deliv. 2018, 25, 1790–1801. [Google Scholar] [CrossRef] [Green Version]
- Iessi, E.; Logozzi, M.; Lugini, L.; Azzarito, T.; Federici, C.; Spugnini, E.P.; Mizzoni, D.; Di Raimo, R.; Angelini, D.F.; Battistini, L.; et al. Acridine orange/exosomes increase the delivery and the effectiveness of acridine orange in human melanoma cells: A new prototype for theranostics of tumors. J. Enzyme Inhib. Med. Chem. 2017, 32, 648–657. [Google Scholar] [CrossRef]
- Zhuang, J.; Tan, J.; Wu, C.; Zhang, J.; Liu, T.; Fan, C.; Li, J.; Zhang, Y. Extracellular vesicles engineered with valency-controlled DNA nanostructures deliver crispr/cas9 system for gene therapy. Nucleic Acids Res. 2020, 48, 8870–8882. [Google Scholar] [CrossRef]
- Ye, Z.; Zhang, T.; He, W.; Jin, H.; Liu, C.; Yang, Z.; Ren, J. Methotrexate-loaded extracellular vesicles functionalized with therapeutic and targeted peptides for the treatment of glioblastoma multiforme. ACS Appl. Mater. Interfaces 2018, 10, 12341–12350. [Google Scholar] [CrossRef]
- Walker, S.; Busatto, S.; Pham, A.; Tian, M.; Suh, A.; Carson, K.; Quintero, A.; Lafrence, M.; Malik, H.; Santana, M.X.; et al. Extracellular vesicle-based drug delivery systems for cancer treatment. Theranostics 2019, 9, 8001–8017. [Google Scholar] [CrossRef] [PubMed]
- Abdollahi, S. Extracellular vesicles from organoids and 3d culture systems. Biotechnol. Bioeng. 2020. [Google Scholar] [CrossRef] [PubMed]
- Pisano, S.; Pierini, I.; Gu, J.; Gazze, A.; Francis, L.W.; Gonzalez, D.; Conlan, R.S.; Corradetti, B. Immune (cell) derived exosome mimetics (idem) as a treatment for ovarian cancer. Front. Cell Dev. Biol. 2020, 8, 553576. [Google Scholar] [CrossRef] [PubMed]
- Yong, T.; Zhang, X.; Bie, N.; Zhang, H.; Zhang, X.; Li, F.; Hakeem, A.; Hu, J.; Gan, L.; Santos, H.A.; et al. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat. Commun. 2019, 10, 3838. [Google Scholar] [CrossRef] [Green Version]
- Taverna, S.; Flugy, A.; Saieva, L.; Kohn, E.C.; Santoro, A.; Meraviglia, S.; De Leo, G.; Alessandro, R. Role of exosomes released by chronic myelogenous leukemia cells in angiogenesis. Int. J. Cancer 2012, 130, 2033–2043. [Google Scholar] [CrossRef] [Green Version]
- Mineo, M.; Garfield, S.H.; Taverna, S.; Flugy, A.; De Leo, G.; Alessandro, R.; Kohn, E.C. Exosomes released by k562 chronic myeloid leukemia cells promote angiogenesis in a src-dependent fashion. Angiogenesis 2012, 15, 33–45. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Wu, X.; Luo, C.; Chen, X.; Yang, L.; Tao, J.; Shi, J. The 786-0 renal cancer cell-derived exosomes promote angiogenesis by downregulating the expression of hepatocyte cell adhesion molecule. Mol. Med. Rep. 2013, 8, 272–276. [Google Scholar] [CrossRef] [Green Version]
- Blache, U.; Horton, E.R.; Xia, T.; Schoof, E.M.; Blicher, L.H.; Schonenberger, A.; Snedeker, J.G.; Martin, I.; Erler, J.T.; Ehrbar, M. Mesenchymal stromal cell activation by breast cancer secretomes in bioengineered 3d microenvironments. Life Sci. Alliance 2019, 2. [Google Scholar] [CrossRef]
- Lugini, L.; Valtieri, M.; Federici, C.; Cecchetti, S.; Meschini, S.; Condello, M.; Signore, M.; Fais, S. Exosomes from human colorectal cancer induce a tumor-like behavior in colonic mesenchymal stromal cells. Oncotarget 2016, 7, 50086–50098. [Google Scholar] [CrossRef] [Green Version]
- Vera, N.; Acuna-Gallardo, S.; Grunenwald, F.; Caceres-Verschae, A.; Realini, O.; Acuna, R.; Lladser, A.; Illanes, S.E.; Varas-Godoy, M. Small extracellular vesicles released from ovarian cancer spheroids in response to cisplatin promote the pro-tumorigenic activity of mesenchymal stem cells. Int. J. Mol. Sci. 2019, 20, 4972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chowdhury, R.; Webber, J.P.; Gurney, M.; Mason, M.D.; Tabi, Z.; Clayton, A. Cancer exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasive myofibroblasts. Oncotarget 2015, 6, 715–731. [Google Scholar] [CrossRef]
- Rai, A.; Greening, D.W.; Xu, R.; Suwakulsiri, W.; Simpson, R.J. Exosomes derived from the human primary colorectal cancer cell line sw480 orchestrate fibroblast-led cancer invasion. Proteomics 2020, 20, e2000016. [Google Scholar] [CrossRef] [PubMed]
- Yoshimura, A.; Sawada, K.; Nakamura, K.; Kinose, Y.; Nakatsuka, E.; Kobayashi, M.; Miyamoto, M.; Ishida, K.; Matsumoto, Y.; Kodama, M.; et al. Exosomal mir-99a-5p is elevated in sera of ovarian cancer patients and promotes cancer cell invasion by increasing fibronectin and vitronectin expression in neighboring peritoneal mesothelial cells. BMC Cancer 2018, 18, 1065. [Google Scholar] [CrossRef]
- Dalchau, N.; Smith, M.J.; Martin, S.; Brown, J.R.; Emmott, S.; Phillips, A. Towards the rational design of synthetic cells with prescribed population dynamics. J. R. Soc. Interface 2012, 9, 2883–2898. [Google Scholar] [CrossRef]
- Cheng, W.C.; Liao, T.T.; Lin, C.C.; Yuan, L.E.; Lan, H.Y.; Lin, H.H.; Teng, H.W.; Chang, H.C.; Lin, C.H.; Yang, C.Y.; et al. Rab27b-activated secretion of stem-like tumor exosomes delivers the biomarker microrna-146a-5p, which promotes tumorigenesis and associates with an immunosuppressive tumor microenvironment in colorectal cancer. Int. J. Cancer 2019, 145, 2209–2224. [Google Scholar] [CrossRef]
- Yeung, V.; Webber, J.P.; Dunlop, E.A.; Morgan, H.; Hutton, J.; Gurney, M.; Jones, E.; Falcon-Perez, J.; Tabi, Z.; Errington, R.; et al. Rab35-dependent extracellular nanovesicles are required for induction of tumour supporting stroma. Nanoscale 2018, 10, 8547–8559. [Google Scholar] [CrossRef] [Green Version]
- Taha, E.A.; Sogawa, C.; Okusha, Y.; Kawai, H.; Oo, M.W.; Elseoudi, A.; Lu, Y.; Nagatsuka, H.; Kubota, S.; Satoh, A.; et al. Knockout of mmp3 weakens solid tumor organoids and cancer extracellular vesicles. Cancers (Basel) 2020, 12, 1260. [Google Scholar] [CrossRef]
- Endzelins, E.; Abols, A.; Buss, A.; Zandberga, E.; Palviainen, M.; Siljander, P.; Line, A. Extracellular vesicles derived from hypoxic colorectal cancer cells confer metastatic phenotype to non-metastatic cancer cells. Anticancer Res. 2018, 38, 5139–5147. [Google Scholar] [CrossRef]
- Yan, Q.; Yuan, W.B.; Sun, X.; Zhang, M.J.; Cen, F.; Zhou, S.Y.; Wu, W.B.; Xu, Y.C.; Tong, L.H.; Ma, Z.H. Asparaginyl endopeptidase enhances pancreatic ductal adenocarcinoma cell invasion in an exosome-dependent manner and correlates with poor prognosis. Int. J. Oncol. 2018, 52, 1651–1660. [Google Scholar] [CrossRef] [Green Version]
- Li, C.; Liu, D.R.; Li, G.G.; Wang, H.H.; Li, X.W.; Zhang, W.; Wu, Y.L.; Chen, L. Cd97 promotes gastric cancer cell proliferation and invasion through exosome-mediated mapk signaling pathway. World J. Gastroenterol. 2015, 21, 6215–6228. [Google Scholar] [CrossRef] [PubMed]
- Guo, D.; Lui, G.Y.L.; Lai, S.L.; Wilmott, J.S.; Tikoo, S.; Jackett, L.A.; Quek, C.; Brown, D.L.; Sharp, D.M.; Kwan, R.Y.Q.; et al. Rab27a promotes melanoma cell invasion and metastasis via regulation of pro-invasive exosomes. Int. J. Cancer 2019, 144, 3070–3085. [Google Scholar] [CrossRef] [Green Version]
- Philip, R.; Heiler, S.; Mu, W.; Buchler, M.W.; Zoller, M.; Thuma, F. Claudin-7 promotes the epithelial-mesenchymal transition in human colorectal cancer. Oncotarget 2015, 6, 2046–2063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Senfter, D.; Holzner, S.; Kalipciyan, M.; Staribacher, A.; Walzl, A.; Huttary, N.; Krieger, S.; Brenner, S.; Jager, W.; Krupitza, G.; et al. Loss of mir-200 family in 5-fluorouracil resistant colon cancer drives lymphendothelial invasiveness in vitro. Hum. Mol. Genet. 2015, 24, 3689–3698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holzner, S.; Senfter, D.; Stadler, S.; Staribacher, A.; Nguyen, C.H.; Gaggl, A.; Geleff, S.; Huttary, N.; Krieger, S.; Jager, W.; et al. Colorectal cancer cell-derived microrna200 modulates the resistance of adjacent blood endothelial barriers in vitro. Oncol. Rep. 2016, 36, 3065–3071. [Google Scholar] [CrossRef] [Green Version]
- Oszvald, A.; Szvicsek, Z.; Papai, M.; Kelemen, A.; Varga, Z.; Tolgyes, T.; Dede, K.; Bursics, A.; Buzas, E.I.; Wiener, Z. Fibroblast-derived extracellular vesicles induce colorectal cancer progression by transmitting amphiregulin. Front. Cell Dev. Biol. 2020, 8, 558. [Google Scholar] [CrossRef]
- Xu, X.; Cheng, J.; Luo, S.; Gong, X.; Huang, D.; Xu, J.; Qian, Y.; Wan, X.; Zhou, H. Deoxycholic acid-stimulated macrophage-derived exosomes promote spasmolytic polypeptide-expressing metaplasia in the stomach. Biochem. Biophys. Res. Commun. 2020, 524, 649–655. [Google Scholar] [CrossRef]
- Li, W.; Han, Y.; Zhao, Z.; Ji, X.; Wang, X.; Jin, J.; Wang, Q.; Guo, X.; Cheng, Z.; Lu, M.; et al. Oral mucosal mesenchymal stem cellderived exosomes: A potential therapeutic target in oral premalignant lesions. Int. J. Oncol. 2019, 54, 1567–1578. [Google Scholar] [CrossRef]
- Rodini, C.O.; Goncalves da Silva, P.B.; Assoni, A.F.; Carvalho, V.M.; Okamoto, O.K. Mesenchymal stem cells enhance tumorigenic properties of human glioblastoma through independent cell-cell communication mechanisms. Oncotarget 2018, 9, 24766–24777. [Google Scholar] [CrossRef] [Green Version]
Model | Advantages | Limitations |
---|---|---|
2D Monolayers | Easy and cost effective Large amount of data available Reproducible cultures, easy to work for downstream applications and imaging | Reduced cell-to-cell interactions Different sensitivity to drugs Loss of biological characteristics over time |
Gel based 3D Cultures | Cell–ECM interactions Possible to incorporate different factors in the gel, extending release over time Uniform spheroids/organoids | Difficult to dispense cells Change of growth media could be irregular Difficult to retrieve cells and downstream analysis |
Low-attachment plates | Simpler and cheaper when compared to gel based systems Long-term culture | Time consuming and low yield achieved Heterogenous spheroids |
Microfluidic systems | Possible chemical gradients Control of fluid rates Convenient for multicellular cultures controlling cell locations | Expensive commercial devices or not well-characterized “in house” build devices Fluidic problems related to bubbles and clogging |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Bordanaba-Florit, G.; Madarieta, I.; Olalde, B.; Falcón-Pérez, J.M.; Royo, F. 3D Cell Cultures as Prospective Models to Study Extracellular Vesicles in Cancer. Cancers 2021, 13, 307. https://doi.org/10.3390/cancers13020307
Bordanaba-Florit G, Madarieta I, Olalde B, Falcón-Pérez JM, Royo F. 3D Cell Cultures as Prospective Models to Study Extracellular Vesicles in Cancer. Cancers. 2021; 13(2):307. https://doi.org/10.3390/cancers13020307
Chicago/Turabian StyleBordanaba-Florit, Guillermo, Iratxe Madarieta, Beatriz Olalde, Juan M. Falcón-Pérez, and Félix Royo. 2021. "3D Cell Cultures as Prospective Models to Study Extracellular Vesicles in Cancer" Cancers 13, no. 2: 307. https://doi.org/10.3390/cancers13020307
APA StyleBordanaba-Florit, G., Madarieta, I., Olalde, B., Falcón-Pérez, J. M., & Royo, F. (2021). 3D Cell Cultures as Prospective Models to Study Extracellular Vesicles in Cancer. Cancers, 13(2), 307. https://doi.org/10.3390/cancers13020307