Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1761 word(s) 1761 2021-12-21 09:23:25 |
2 The format is correct. Meta information modification 1761 2021-12-22 02:15:30 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Tominaga, N. Anti-Cancer Role and Therapeutic Potential of Extracellular Vesicles. Encyclopedia. Available online: https://encyclopedia.pub/entry/17376 (accessed on 01 July 2024).
Tominaga N. Anti-Cancer Role and Therapeutic Potential of Extracellular Vesicles. Encyclopedia. Available at: https://encyclopedia.pub/entry/17376. Accessed July 01, 2024.
Tominaga, Naoomi. "Anti-Cancer Role and Therapeutic Potential of Extracellular Vesicles" Encyclopedia, https://encyclopedia.pub/entry/17376 (accessed July 01, 2024).
Tominaga, N. (2021, December 21). Anti-Cancer Role and Therapeutic Potential of Extracellular Vesicles. In Encyclopedia. https://encyclopedia.pub/entry/17376
Tominaga, Naoomi. "Anti-Cancer Role and Therapeutic Potential of Extracellular Vesicles." Encyclopedia. Web. 21 December, 2021.
Anti-Cancer Role and Therapeutic Potential of Extracellular Vesicles
Edit

Cell–cell communication is an important mechanism in biological processes. Extracellular vesicles (EVs), also referred to as exosomes, microvesicles, and prostasomes, are microvesicles secreted from a variety of cells. Importantly, EVs contribute to cancer malignancy mechanisms such as carcinogenesis, proliferation, angiogenesis, metastasis, and escape from the immune system. As EVs are thought to be secreted into body fluids, they have the potential to serve as diagnostic markers for liquid biopsy. In addition, the characteristics of EVs make them suitable for use in drug delivery systems and novel cancer treatments. 

cancer extracellular vesicles exosome cancer therapy

1. Introduction

Extracellular vesicles (EVs), which also go by exosomes, microvesicles, and prostasomes, are secreted from a variety of cells [1][2] (Table 1). EVs are nanometer-scale vesicles composed of a lipid bilayer and contain biologically functional molecules, such as microRNAs (miRNAs), mRNAs, and proteins [3]. In recent years, EVs have been recognized as a cell–cell communication tool. The basic idea of cell–cell communication using EVs is that EVs secreted from donor cells are taken up by recipient cells in a paracrine or autocrine manner. EVs modify the condition of recipient cells by the biological molecules contained within them. The history of EVs dates back approximately 40 years, when Hans Lutz et al. reported the release of vesicles from old sheep erythrocytes [4]. Ronquist reported a functional fraction in the supernatant of prostatic fluid in the same year [5]. It has been reported that red blood cells secrete vesicles containing proteins and lipids during maturation [6][7][8]. Some research groups have reported that the transferrin receptor is internalized in vesicles made by multivesicular bodies [9][10][11]. However, in 1991, Johnstone et al. concluded that vesicles secreted from cells were “a garbage bin” for unnecessary membrane proteins [12]. Importantly, the vesicles secreted from cells contain mRNA and miRNA, which can be transferred to other cells and be functional in them [13].
Table 1. The features of extracellular vesicles.
  Extracellular Vesicles (EVs)
Terminology Exosomes Microvesicles Apoptotic Bodies
Origin Multivesicle body Plasma membrane Plasma membrane
Size 50–150 nm 100–1000 nm 100–5000 nm
Marker proteins CD9, CD63, Tsg101 etc. Integrins, Selectins, CD40 etc. Annexin V, thrombospondin, C3b etc.
References [1][3] [1][14] [1][15]

In the past two decades, EVs have been shown to play a crucial role in cancer biology. Accumulating evidence indicates the importance of cell–cell communication through EVs in cancer malignancy mechanisms, such as cancer cell proliferation [16], immune modulation [17], angiogenesis [18], metastasis [19], and pro-metastasis niche formation [20] (Figure 1). Importantly, in the early history of EV research, EVs derived from dendritic cells (DCs) pulsed with tumor peptides were proposed as a cell-free vaccine method [21]. Furthermore, a potential cancer therapeutic strategy based on the suppression of cancer metastasis via the removal of EVs that contribute to cancer malignancy has been reported [22].

Figure 1. EVs contribute to cancer malignancy and they have an emerging role of therapeutic potential in cancer malignancy.

2. Carcinogenesis

Cancer cells emerge from cells damaged by various factors, such as inflammation, chemical stress, radiation, oxidative stress, and aging [23]. These stresses, especially aging, affect the formation of malignant tumors through the accumulation of genetic and epigenetic changes in genes. It is likely that there is a relationship between these causes of carcinogenesis. Chronic inflammation, such as “inflammaging,” is a risk factor for carcinogenesis, and is caused by cytokines and chemokines [24]. It is predicted that elderly people probably have chronic inflammation without infection caused by senescent cells. The senescence-associated secretory phenotype is a feature of senescent cells that leads to chronic inflammation in elderly people, with factors such as interleukin (IL) -6, and IL-8 secreted by senescent cells [25]. EVs are also secreted from senescent cells, and they may exert detrimental effects [26][27]. Accumulating evidence suggests that EVs contribute to carcinogenesis or precancerous conditions, such as inflammation [28][29][30][31], fibrosis [32][33][34], double-strand breaks in DNA [35][34], and endoplasmic reticulum (ER) stress [36].
Bladder cancer cell-derived EVs induce neoplastic transformation of nonmalignant cells through the induction of the unfolded protein response in the ER [36]. EVs have been suggested to affect tumor recurrence and the potential for carcinogenesis. The Epstein–Barr virus M81-infected B cells release EVs that contain non-coding Epstein–Barr virus-encoded RNA, which are then taken up by B cells [28], which results in chronic inflammation. Inflammation and carcinogenesis caused by viral infection may be linked. EVs derived from macrophages have been reported to upregulate TBC1 domain family member 3 by downregulating stanniocalcin-1-mediated inflammation [29]

3. Proliferation

“Enabling replicative immortality” is a hallmark of cancer [37]. HeLa cells were established in 1953 and contribute greatly to cancer research because of their proliferation on a dish [38]. It is well known that activation of telomerase is one of the causes of proliferation, as it is not subject to the Hayflick limit. Uncontrolled proliferation is a fundamental feature of cancer that leads to gene mutations, metabolic changes, and epigenetic alterations possibly resulting in malignancy. Cancer cell-derived EVs contribute to cell proliferation and growth [39][40][41][42][43].
EVs from mesenchymal stem cells (MSCs) play a dual role in cancer biology. They exhibit potential as anti-cancer treatments but also contribute to cancer malignancy [39][44]. Importantly, MSCs are educated by cancer-derived EVs to contribute to cancer malignancy. It has been reported that EVs derived from cholangiocarcinoma-educated bone marrow MSCs enhance the secretion of C-X-C motif chemokine ligand (CXCL)-1, C-C motif chemokine ligand 2 (CCL2), and IL-6, which affect cancer proliferation [39]. MiR-410 containing EVs derived from human umbilical cord MSCs decreases phosphatase and tensin homolog (PTEN) expression in lung adenocarcinoma. These results suggests that the uptake of EVs by lung adenocarcinoma increases proliferation and decreases apoptosis. Cancer-associated fibroblasts (CAFs) also play a crucial role in cancer proliferation [45]

4. Angiogenesis and Intravasation

Angiogenesis and lymphangiogenesis are important for the survival and progression of cancer cells, which are activated by signals from cancer cells that are growing [46]. These are important steps for the supply of oxygen, nutrients, and metabolism in cancer cells [47]. Vascular endothelial growth factor (VEGF), basic fibroblast growth factor, angiogenin, and TGF-α play an important role in angiogenesis [47]. Cancer-derived EVs also play an important role in angiogenesis [42][48][49][50][51][52][53][54] and lymph-angiogenesis [55]. EVs derived from colorectal cancer cells activate early growth response protein-1 in endothelial cells, causing the migration of endothelial cells [56]. Cancer-derived EVs stimulate MSCs to form vessel-like formations [57]. It has been reported that miRNAs in EVs also plays an important role in angiogenesis [58]. Rac1-, PAK2- [59], VEGF- [60], and angiopoietin-2-containing [61] EVs are related to angiogenesis. These results suggest that cancer-derived EVs promote angiogenesis. Lymph nodes are a route of cancer metastasis [62]. It is reported that laminin γ2-containing EVs promote lymphangiogenesis [63]; however, mechanisms underlying lymphatic intravasation remain unclear.

5. Metastasis

Cancer cells can metastasize to any part of the body; however, sites such as bone, the liver, and the lungs are the most common. Brain metastasis is a critical cause of death. Uncontrolled cancer metastasis is a major cause of cancer-related deaths. Cancer metastasis involves multiple steps, such as epithelial–mesenchymal transition (EMT), intravasation, extravasation, and proliferation at the metastatic organ. The seed-and-soil theory is well accepted as a mechanism of metastasis [64]. Using the metastatic efficiency index, Weiss suggested that 65% of metastasis seems to be caused by the amount of organ blood flow [64][65]. In contrast, there are common sites of cancer metastasis and sites that are specific to the cancer type. In such complicated mechanisms of metastasis, EVs contribute to EMT [66][67][68], migration [69][70][71][72], metastasis niche formation [20][73][74], metastasis promotion [73], and the tumor microenvironment [75].

6. Escape from Immune System

The immune system consists of macrophages, B cells, T cells, and DCs. It protects the body from invaders such as bacteria, viruses, and toxins; contributes to the recovery of the body; and removes cancer cells. However, cancer cells can escape the immune system by modifying them through EVs. Programmed death ligand-1 (PD-L1) is a receptor that suppresses or stops T-cell reactions by binding to programmed cell death-1 (PD-1). EVs derived from glioblastoma promote immune evasion through PD-1 binding to PD-L1 on EVs [76][77]. Ovarian cancer cell-derived EVs have been reported to inhibit T-cell receptor-dependent activation in T-cells [78]. EVs derived from tumor-associated macrophages have immunosuppressive effects; conversely, these cells have the potential to activate anti-tumor immunity [79]. MSC-derived EVs containing miR-222 contribute to immune escape in colorectal cancer by downregulating the AKT pathway [80]. These reports suggest that EVs derived from cancer cells and cells in the microenvironment suppress the immune system.

7. Chemotherapeutic Stress

Bone marrow MSC-derived EVs increase the viability of multiple myeloma cells and drug resistance [81]. Non-small-cell lung cancer cell-derived EVs increase gefitinib-induced apoptosis [82]. In EVs, lncRNAs contribute to drug resistance [83][84][85][86][87][88], with lncRNA H19 in EVs increasing gefitinib resistance in non-small-cell lung cancer cells [89]. EVs derived from MSCs increase drug resistance [72]. MSC-EVs are collected under stress using a non-serum culture medium. EVs derived from under-stressed MSCs decrease doxorubicin-induced apoptosis in osteosarcoma cells, and those derived from melanoma cells containing anaplastic lymphoma kinase could transfer drug resistance to other melanoma cells [90]. EVs derived from tumor-associated macrophages, which are components of the cancer microenvironment, increase resistance to the anticancer drug gemcitabine [91]. MiR-30b-3p in EVs derived from hypoxic glioma cells contributes to drug resistance by decreasing the expression of ras homolog family member B [92].

8. Potential of EVs for Liquid Biopsy

EVs secreted into the extracellular environment may be related to mechanisms of cancer malignancy. Many reports have suggested that EVs contain specific molecules that contribute to cancer malignancy or related cancer types. Therefore, it is possible that EVs can be used for diagnosis. A liquid biopsy is a test that uses body fluids such as blood, bone marrow, saliva, urine, and tears. A minimally invasive method is required in liquid biopsy as much as possible to avoid pain. Bone marrow biopsy is an invasive method, as is blood biopsy. On the other hand, saliva, urine, and tears are non-invasive methods of biopsy. Liquid biopsy tests can identify early stage, progression, and metastasis of cancer by detecting specific molecules. The idea of a liquid biopsy using EVs is to detect cancer-specific EV molecules such as miRNAs, DNAs, and proteins for the diagnosis of cancer [93]

9. Potential of EVs for Cancer Treatment

The idea of cancer treatment using EVs has three aspects—(1) inhibition of EV production, (2) disruption of EV uptake, and (3) elimination of EVs [94]. In addition, there is a strategy for the application of EVs that carry a druggable molecule for cancer treatment [95]. CD63 and CD9 are used as markers for targeting EVs in the circulation. EV elimination from the circulation using anti-CD63 and CD9 antibodies can reduce cancer metastasis [22]. The CD9 Fab fragment also inhibits EV internalization [96]. These reports suggest that targeting proteins on EVs can be used for the neutralization and elimination of EVs from the blood. It has also been suggested that potential therapeutics exert anti-cancer effects by blocking EVs containing TGF-β [97]; however, the therapeutics strategy of “inhibition of EV production” is still unclear. 

References

  1. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17.
  2. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383.
  3. Yáñez-Mó, M.; Siljander, P.R.M.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066.
  4. Lutz, H.U.; Lomant, A.J.; McMillan, P.; Wehrli, E. Rearrangements of integral membrane components during in vitro aging of sheep erythrocyte membranes. J. Cell Biol. 1977, 74, 389–398.
  5. Ronquist, G.; Hedström, M. Restoration of detergent-inactivated adenosine triphosphatase activity of human prostatic fluid with concanavalin A. Biochim. Biophys. Acta. 1977, 483, 483–486. Available online: https://www.sciencedirect.com/science/article/pii/000527447790078X?via%3Dihub (accessed on 15 December 2021).
  6. Müller, H.; Schmidt, U.; Lutz, H.U. On the mechanism of vesicle release from ATP-depleted human red blood cells. Biochim. Biophys. Acta. 1981, 649, 462–470.
  7. Zweig, S.E.; Tokuyasu, K.T.; Singer, S.J. Member-associated changes during erythropoiesis. On the mechanism of maturation of reticulocytes to erythrocytes. J. Supramol. Struct. Cell. Biochem. 1981, 17, 163–181.
  8. Pan, B.T.; Johnstone, R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 1983, 33, 967–978.
  9. Harding, C.; Heuser, J.; Stahl, P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J. Cell Biol. 1983, 97, 329–339.
  10. Harding, C.; Heuser, J.; Stahl, P. Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: Demonstration of a pathway for receptor shedding. Eur. J. Cell Biol. 1984, 35, 256–263. Available online: https://www.ncbi.nlm.nih.gov/pubmed/?term=6151502%5Buid%5D. (accessed on 15 December 2021).
  11. Pan, B.T.; Teng, K.; Wu, C.; Adam, M.; Johnstone, R.M. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J. Cell Biol. 1985, 101, 942–948.
  12. Johnstone, R.M.; Mathew, A.; Mason, A.B.; Teng, K. Exosome formation during maturation of mammalian and avian reticulocytes: Evidence that exosome release is a major route for externalization of obsolete membrane proteins. J. Cell. Physiol. 1991, 147, 27–36.
  13. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659.
  14. Camussi, G.; Deregibus, M.-C.; Bruno, S.; Grange, C.; Fonsato, V.; Tetta, C. Exosome/microvesicle-mediated epigenetic reprogramming of cells. Am. J. Cancer Res. 2011, 1, 98–110.
  15. Akers, J.C.; Gonda, D.; Kim, R.; Carter, B.S.; Chen, C.C. Biogenesis of extracellular vesicles (EV): Exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J. Neurooncol. 2013, 113, 1–11.
  16. Al-Nedawi, K.; Meehan, B.; Micallef, J.; Lhotak, V.; May, L.; Guha, A.; Rak, J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat. Cell Biol. 2008, 10, 619–624.
  17. Clayton, A.; Tabi, Z. Exosomes and the MICA-NKG2D system in cancer. Blood Cells. Mol. Dis. 2005, 34, 206–213.
  18. Kosaka, N.; Iguchi, H.; Hagiwara, K.; Yoshioka, Y.; Takeshita, F.; Ochiya, T. Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic micrornas regulate cancer cell metastasis. J. Biol. Chem. 2013, 288, 10849–10859.
  19. Tominaga, N.; Kosaka, N.; Ono, M.; Katsuda, T.; Yoshioka, Y.; Tamura, K.; Lötvall, J.; Nakagama, H.; Ochiya, T. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier. Nat. Commun. 2015, 6, 6716.
  20. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.M.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891.
  21. Zitvogel, L.; Regnault, A.; Lozier, A.; Wolfers, J.; Flament, C.; Tenza, D.; Ricciardi-Castagnoli, P.; Raposo, G.; Amigorena, S. Eradication of established murine tumors using a novel cell-free vaccine: Dendritic cell-derived exosomes. Nat. Med. 1998, 4, 594–600.
  22. Nishida-Aoki, N.; Tominaga, N.; Takeshita, F.; Sonoda, H.; Yoshioka, Y.; Ochiya, T. Disruption of Circulating Extracellular Vesicles as a Novel Therapeutic Strategy against Cancer Metastasis. Mol. Ther. 2017, 25, 181–191.
  23. Valavanidis, A.; Vlachogianni, T.; Fiotakis, K.; Loridas, S. Pulmonary oxidative stress, inflammation and cancer: Respirable particulate matter, fibrous dusts and ozone as major causes of lung carcinogenesis through reactive oxygen species mechanisms. Int. J. Environ. Res. Public Health 2013, 10, 3886–3907.
  24. Franceschi, C.; Campisi, J. Chronic inflammation (Inflammaging) and its potential contribution to age-associated diseases. J.Gerontol. Ser. A Biol. Sci. Med. Sci. 2014, 69, S4–S9.
  25. Coppé, J.-P.; Patil, C.K.; Rodier, F.; Sun, Y.; Muñoz, D.P.; Goldstein, J.; Nelson, P.S.; Desprez, P.-Y.; Campisi, J. Senescence-Associated Secretory Phenotypes Reveal Cell-Nonautonomous Functions of Oncogenic RAS and the p53 Tumor Suppressor. PLoS Biol. 2008, 6, e301.
  26. Loarca, L.; de Assuncao, T.M.; Jalan-Sakrikar, N.; Bronk, S.; Krishnan, A.; Huang, B.; Morton, L.; Trussoni, C.; Bonilla, L.M.; Krueger, E.; et al. Development and characterization of cholangioids from normal and diseased human cholangiocytes as an in vitro model to study primary sclerosing cholangitis. Lab. Investig. 2017, 97, 1385–1396.
  27. Jakhar, R.; Crasta, K. Exosomes as emerging pro-tumorigenic mediators of the senescence-associated secretory phenotype. Int. J. Mol. Sci. 2019, 20, 2547.
  28. Li, Z.; Tsai, M.-H.; Shumilov, A.; Baccianti, F.; Tsao, S.W.; Poirey, R.; Delecluse, H.-J. Epstein-Barr virus ncRNA from a nasopharyngeal carcinoma induces an inflammatory response that promotes virus production. Nat. Microbiol. 2019, 4, 2475–2486.
  29. Leung, C.C.T.; Wong, C.K.C. Characterization of stanniocalcin-1 expression in macrophage differentiation. Transl. Oncol. 2021, 14, 100881.
  30. Chaiyadet, S.; Sotillo, J.; Smout, M.; Cantacessi, C.; Jones, M.K.; Johnson, M.S.; Turnbull, L.; Whitchurch, C.B.; Potriquet, J.; Laohaviroj, M.; et al. Carcinogenic liver fluke secretes extracellular vesicles that promote cholangiocytes to adopt a tumorigenic phenotype. J. Infect. Dis. 2015, 212, 1636–1645.
  31. Iuliano, M.; Mangino, G.; Chiantore, M.V.; Zangrillo, M.S.; Accardi, R.; Tommasino, M.; Fiorucci, G.; Romeo, G. Human Papillomavirus E6 and E7 oncoproteins affect the cell microenvironment by classical secretion and extracellular vesicles delivery of inflammatory mediators. Cytokine 2018, 106, 182–189.
  32. Matsuura, K.; de Giorgi, V.; Schechterly, C.; Wang, R.Y.; Farci, P.; Tanaka, Y.; Alter, H.J. Circulating let-7 levels in plasma and extracellular vesicles correlate with hepatic fibrosis progression in chronic hepatitis C. Hepatology 2016, 64, 732–745.
  33. Kim, J.H.; Lee, C.H.; Lee, S.-W. Exosomal Transmission of MicroRNA from HCV Replicating Cells Stimulates Transdifferentiation in Hepatic Stellate Cells. Mol. Ther. Nucleic Acids 2019, 14, 483–497.
  34. Zhang, X.; Deeke, S.A.; Ning, Z.; Starr, A.E.; Butcher, J.; Li, J.; Mayne, J.; Cheng, K.; Liao, B.; Li, L.; et al. Metaproteomics reveals associations between microbiome and intestinal extracellular vesicle proteins in pediatric inflammatory bowel disease. Nat. Commun. 2018, 9, 1–14.
  35. Butin-Israeli, V.; Bui, T.M.; Wiesolek, H.L.; Mascarenhas, L.; Lee, J.J.; Mehl, L.C.; Knutson, K.R.; Adam, S.A.; Goldman, R.D.; Beyder, A.; et al. Neutrophil-induced genomic instability impedes resolution of inflammation and wound healing. J. Clin. Investig. 2019, 129, 712–726.
  36. Wu, C.H.; Silvers, C.R.; Messing, E.M.; Lee, Y.F. Bladder cancer extracellular vesicles drive tumorigenesis by inducing the unfolded protein response in endoplasmic reticulum of nonmalignant cells. J. Biol. Chem. 2019, 294, 3207–3218.
  37. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674.
  38. Scherer, W.F.; Syverton, J.T.; Gey, G.O. Studies on the propagation in vitro of poliomyelitis viruses. IV. Viral multiplication in a stable strain of human malignant epithelial cells (strain HeLa) derived from an epidermoid carcinoma of the cervix. J. Exp. Med. 1953, 97, 695–710.
  39. Haga, H.; Yan, I.K.; Takahashi, K.; Wood, J.; Zubair, A.; Patel, T. Tumour cell-derived extracellular vesicles interact with mesenchymal stem cells to modulate the microenvironment and enhance cholangiocarcinoma growth. J. Extracell. Vesicles 2015, 4, 24900.
  40. Zhou, X.; Li, T.; Chen, Y.; Zhang, N.; Wang, P.; Liang, Y.; Long, M.; Liu, H.; Mao, J.; Liu, Q.; et al. Mesenchymal stem cell-derived extracellular vesicles promote the in vitro proliferation and migration of breast cancer cells through the activation of the ERK pathway. Int. J. Oncol. 2019, 54, 1843–1852.
  41. Czystowska-Kuzmicz, M.; Sosnowska, A.; Nowis, D.; Ramji, K.; Szajnik, M.; Chlebowska-Tuz, J.; Wolinska, E.; Gaj, P.; Grazul, M.; Pilch, Z.; et al. Small extracellular vesicles containing arginase-1 suppress T-cell responses and promote tumor growth in ovarian carcinoma. Nat. Commun. 2019, 10, 3000.
  42. Tsutsui, T.; Kawahara, H.; Kimura, R.; Dong, Y.; Jiapaer, S.; Sabit, H.; Zhang, J.; Yoshida, T.; Nakada, M.; Hanayama, R. Glioma-derived extracellular vesicles promote tumor progression by conveying WT1. Carcinogenesis 2020, 41, 1238–1245.
  43. Li, J.; Xue, J.; Ling, M.; Sun, J.; Xiao, T.; Dai, X.; Sun, Q.; Cheng, C.; Xia, H.; Wei, Y.; et al. MicroRNA-15b in extracellular vesicles from arsenite-treated macrophages promotes the progression of hepatocellular carcinomas by blocking the LATS1-mediated Hippo pathway. Cancer Lett. 2021, 497, 137–153.
  44. Dong, L.; Pu, Y.; Zhang, L.; Qi, Q.; Xu, L.; Li, W.; Wei, C.; Wang, X.; Zhou, S.; Zhu, J.; et al. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles promote lung adenocarcinoma growth by transferring miR-410. Cell Death Dis. 2018, 9, 218.
  45. Richards, K.E.; Zeleniak, A.E.; Fishel, M.L.; Wu, J.; Littlepage, L.E.; Hill, R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2017, 36, 1770–1778.
  46. Folkman, J. Tumor angiogenesis: Therapeutic implications. N. Engl. J. Med. 1971, 285, 1182–1186.
  47. Nishida, N.; Yano, H.; Nishida, T.; Kamura, T.; Kojiro, M. Angiogenesis in cancer. Vasc. Health Risk Manag. 2006, 2, 213–219.
  48. Baj-Krzyworzeka, M.; Mytar, B.; Weglarczyk, K.; Szatanek, R.; Kijowski, J.; Siedlar, M. Protumorogenic Potential of Pancreatic Adenocarcinoma-Derived Extracellular Vesicles. Folia Biol. 2020, 66, 104–110.
  49. De Andrade, A.; de Oliveira, C.E.; Dourado, M.R.; Macedo, C.; Winck, F.V.; Leme, A.F.P.; Salo, T.; Coletta, R.D.; Freitas, R.d.; Galvão, H.C. Extracellular vesicles from oral squamous carcinoma cells display pro- and anti-angiogenic properties. Oral Dis. 2018, 24, 725–731.
  50. Ren, J.G.; Zhang, W.; Liu, B.; Man, Q.W.; Xiong, X.P.; Li, C.; Zhu, J.Y.; Wang, W.M.; Jia, J.; Sun, Z.J.; et al. Clinical Significance and Roles in Angiogenesis of Circulating Microparticles in Oral Cancer. J. Dent. Res. 2016, 95, 860–867.
  51. Zarfati, M.; Avivi, I.; Brenner, B.; Katz, T.; Aharon, A. Extracellular vesicles of multiple myeloma cells utilize the proteasome inhibitor mechanism to moderate endothelial angiogenesis. Angiogenesis 2019, 22, 185–196.
  52. Lucero, R.; Zappulli, V.; Sammarco, A.; Murillo, O.D.; Cheah, P.S.; Srinivasan, S.; Tai, E.; Ting, D.T.; Wei, Z.; Roth, M.E.; et al. Glioma-Derived miRNA-Containing Extracellular Vesicles Induce Angiogenesis by Reprogramming Brain Endothelial Cells. Cell Rep. 2020, 30, 2065–2074.e4.
  53. Li, J.; Liu, X.; Zang, S.; Zhou, J.; Zhang, F.; Sun, B.; Qi, D.; Li, X.; Kong, J.; Jin, D.; et al. Small extracellular vesicle-bound vascular endothelial growth factor secreted by carcinoma-associated fibroblasts promotes angiogenesis in a bevacizumab-resistant manner. Cancer Lett. 2020, 492, 71–83.
  54. Garcia-Hernandez, A.; Leal-Orta, E.; Ramirez-Ricardo, J.; Cortes-Reynosa, P.; Thompson-Bonilla, R.; Salazar, E.P. Linoleic acid induces secretion of extracellular vesicles from MDA-MB-231 breast cancer cells that mediate cellular processes involved with angiogenesis in HUVECs. Prostaglandins Other Lipid Mediat. 2021, 153, 106519.
  55. Carrasco-Ramírez, P.; Greening, D.W.; Andrés, G.; Gopal, S.K.; Martín-Villar, E.; Renart, J.; Simpson, R.J.; Quintanilla, M. Podoplanin is a component of extracellular vesicles that reprograms cell-derived exosomal proteins and modulates lymphatic vessel formation. Oncotarget 2016, 7, 16070–16089.
  56. Yoon, Y.J.; Kim, D.-K.; Yoon, C.M.; Park, J.; Kim, Y.-K.; Roh, T.-Y.; Gho, Y.S. Egr-1 activation by cancer-derived extracellular vesicles promotes endothelial cell migration via ERK1/2 and JNK signaling pathways. PLoS ONE 2014, 9, e115170.
  57. Lindoso, R.S.; Collino, F.; Camussi, G. Extracellular vesicles derived from renal cancer stem cells induce a pro-tumorigenic phenotype in mesenchymal stromal cells. Oncotarget 2015, 6, 7959–7969.
  58. Shao, Z.; Pan, Q.; Zhang, Y. Hepatocellular carcinoma cell-derived extracellular vesicles encapsulated microRNA-584-5p facilitates angiogenesis through PCK1-mediated nuclear factor E2-related factor 2 signaling pathway. Int. J. Biochem. Cell Biol. 2020, 125, 105789.
  59. Gopal, S.K.; Greening, D.W.; Hanssen, E.G.; Zhu, H.-J.; Simpson, R.J.; Mathias, R.A. Oncogenic epithelial cell-derived exosomes containing Rac1 and PAK2 induce angiogenesis in recipient endothelial cells. Oncotarget 2016, 7, 19709–19722.
  60. Feng, Q.; Zhang, C.; Lum, D.; Druso, J.E.; Blank, B.; Wilson, K.F.; Welm, A.; Antonyak, M.A.; Cerione, R.A. A class of extracellular vesicles from breast cancer cells activates VEGF receptors and tumour angiogenesis. Nat. Commun. 2017, 8, 14450.
  61. Xie, J.-Y.; Wei, J.-X.; Lv, L.-H.; Han, Q.-F.; Yang, W.-B.; Li, G.-L.; Wang, P.-X.; Wu, S.-B.; Duan, J.-X.; Zhuo, W.-F.; et al. Angiopoietin-2 induces angiogenesis via exosomes in human hepatocellular carcinoma. Cell Commun. Signal. 2020, 18, 46.
  62. Weigelt, B.; Peterse, J.L.; Veer, L.J.v. Breast cancer metastasis: Markers and models. Nat. Rev. Cancer 2005, 5, 591–602.
  63. Wang, S.-H.; Liou, G.-G.; Liu, S.-H.; Chang, J.S.; Hsiao, J.-R.; Yen, Y.-C.; Chen, Y.-L.; Wu, W.-L.; Chang, J.-Y.; Chen, Y.-W. Laminin γ2-enriched extracellular vesicles of oral squamous cell carcinoma cells enhance in vitro lymphangiogenesis via integrin α3-dependent uptake by lymphatic endothelial cells. Int. J. Cancer 2019, 144, 2795–2810.
  64. Tominaga, N.; Katsuda, T.; Ochiya, T. Micromanaging of tumor metastasis by extracellular vesicles. Semin. Cell Dev. Biol. 2015, 40, 52–59.
  65. Weiss, L. Comments on hematogenous metastatic patterns in humans as revealed by autopsy. Clin. Exp. Metastasis 1992, 10, 191–199.
  66. Galindo-Hernandez, O.; Serna-Marquez, N.; Castillo-Sanchez, R.; Salazar, E.P. Extracellular vesicles from MDA-MB-231 breast cancer cells stimulated with linoleic acid promote an EMT-like process in MCF10A cells. Prostaglandins. Leukot. Essent. Fatty Acids 2014, 91, 299–310.
  67. Schweiger, M.W.; Li, M.; Giovanazzi, A.; Fleming, R.L.; Tabet, E.I.; Nakano, I.; Würdinger, T.; Chiocca, E.A.; Tian, T.; Tannous, B.A. Extracellular Vesicles Induce Mesenchymal Transition and Therapeutic Resistance in Glioblastomas through NF-κB/STAT3 Signaling. Adv. Biosyst. 2020, 4, e1900312.
  68. Lucchetti, D.; Perelli, L.; Colella, F.; Ricciardi-Tenore, C.; Scoarughi, G.L.; Barbato, G.; Boninsegna, A.; de Maria, R.; Sgambato, A. Low-intensity pulsed ultrasound affects growth, differentiation, migration, and epithelial-to-mesenchymal transition of colorectal cancer cells. J. Cell. Physiol. 2020, 235, 5363–5377.
  69. Liu, C.; Zhou, X.; Long, Q.; Zeng, H.; Sun, Q.; Chen, Y.; Wu, D.; Liu, L. Small extracellular vesicles containing miR-30a-3p attenuate the migration and invasion of hepatocellular carcinoma by targeting SNAP23 gene. Oncogene 2021, 40, 233–245.
  70. Chiba, M.; Watanabe, N.; Watanabe, M.; Sakamoto, M.; Sato, A.; Fujisaki, M.; Kubota, S.; Monzen, S.; Maruyama, A.; Nanashima, N.; et al. Exosomes derived from SW480 colorectal cancer cells promote cell migration in HepG2 hepatocellular cancer cells via the mitogen-activated protein kinase pathway. Int. J. Oncol. 2016, 48, 305–312.
  71. Lopatina, T.; Favaro, E.; Danilova, L.; Fertig, E.J.; Favorov, A.V.; Kagohara, L.T.; Martone, T.; Bussolati, B.; Romagnoli, R.; Albera, R.; et al. Extracellular Vesicles Released by Tumor Endothelial Cells Spread Immunosuppressive and Transforming Signals Through Various Recipient Cells. Front. Cell Dev. Biol. 2020, 8, 698.
  72. Vallabhaneni, K.C.; Hassler, M.-Y.; Abraham, A.; Whitt, J.; Mo, Y.-Y.; Atfi, A.; Pochampally, R. Mesenchymal Stem/Stromal Cells under Stress Increase Osteosarcoma Migration and Apoptosis Resistance via Extracellular Vesicle Mediated Communication. PLoS ONE 2016, 11, e0166027.
  73. Macklin, R.; Wang, H.; Loo, D.; Martin, S.; Cumming, A.; Cai, N.; Lane, R.; Ponce, N.S.; Topkas, E.; Inder, K.; et al. Extracellular vesicles secreted by highly metastatic clonal variants of osteosarcoma preferentially localize to the lungs and induce metastatic behaviour in poorly metastatic clones. Oncotarget 2016, 7, 43570–43587.
  74. 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.
  75. Dörsam, B.; Bösl, T.; Reiners, K.S.; Barnert, S.; Schubert, R.; Shatnyeva, O.; Zigrino, P.; Engert, A.; Hansen, H.P.; von Strandmann, E.P. Hodgkin Lymphoma-Derived Extracellular Vesicles Change the Secretome of Fibroblasts Toward a CAF Phenotype. Front. Immunol. 2018, 9, 1358.
  76. Ricklefs, F.L.; Alayo, Q.; Krenzlin, H.; Mahmoud, A.B.; Speranza, M.C.; Nakashima, H.; Hayes, J.L.; Lee, K.; Balaj, L.; Passaro, C.; et al. Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci. Adv. 2018, 4, eaar2766.
  77. Himes, B.T.; Peterson, T.E.; de Mooij, T.; Garcia, L.M.C.; Jung, M.-Y.; Uhm, S.; Yan, D.; Tyson, J.; Jin-Lee, H.J.; Parney, D.; et al. The role of extracellular vesicles and PD-L1 in glioblastoma-mediated immunosuppressive monocyte induction. Neuro-Oncology 2020, 22, 967–978.
  78. Shenoy, G.N.; Loyall, J.; Maguire, O.; Iyer, V.; Kelleher, R.J.J.; Minderman, H.; Wallace, P.K.; Odunsi, K.; Balu-Iyer, S.V.; Bankert, R.B. Exosomes Associated with Human Ovarian Tumors Harbor a Reversible Checkpoint of T-cell Responses. Cancer Immunol. Res. 2018, 6, 236–247.
  79. Cianciaruso, C.; Beltraminelli, T.; Duval, F.; Nassiri, S.; Hamelin, R.; Mozes, A.; Gallart-Ayala, H.; Torres, G.C.; Torchia, B.; Ries, C.H.; et al. Molecular Profiling and Functional Analysis of Macrophage-Derived Tumor Extracellular Vesicles. Cell Rep. 2019, 27, 3062–3080.e11.
  80. Li, S.; Yan, G.; Yue, M.; Wang, L. Extracellular vesicles-derived microRNA-222 promotes immune escape via interacting with ATF3 to regulate AKT1 transcription in colorectal cancer. BMC Cancer 2021, 21, 349.
  81. Wang, J.; Hendrix, A.; Hernot, S.; Lemaire, M.; de Bruyne, E.; van Valckenborgh, E.; Lahoutte, T.; de Wever, O.; Vanderkerken, K.; Menu, E. Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood 2014, 124, 555–566.
  82. Choi, D.-Y.; You, S.; Jung, J.H.; Lee, J.C.; Rho, J.K.; Lee, K.Y.; Freeman, M.R.; Kim, K.P.; Kim, J. Extracellular vesicles shed from gefitinib-resistant nonsmall cell lung cancer regulate the tumor microenvironment. Proteomics 2014, 14, 1845–1856.
  83. Takahashi, K.; Yan, I.K.; Wood, J.; Haga, H.; Patel, T. Involvement of extracellular vesicle long noncoding RNA (linc-VLDLR) in tumor cell responses to chemotherapy. Mol. Cancer Res. 2014, 12, 1377–1387.
  84. Chen, Y.; Liu, L.; Li, J.; Du, Y.; Wang, J.; Liu, J. Effects of long noncoding RNA (linc-VLDLR) existing in extracellular vesicles on the occurrence and multidrug resistance of esophageal cancer cells. Pathol. Res. Pract. 2019, 215, 470–477.
  85. Chen, X.; Liu, Y.; Zhang, Q.; Liu, B.; Cheng, Y.; Zhang, Y.; Sun, Y.; Liu, J.; Gen, H. Exosomal Long Non-coding RNA HOTTIP Increases Resistance of Colorectal Cancer Cells to Mitomycin via Impairing MiR-214-Mediated Degradation of KPNA3. Front. Cell Dev. Biol. 2020, 8, 582723.
  86. Zhang, W.; Cai, X.; Yu, J.; Lu, X.; Qian, Q.; Qian, W. Exosome-mediated transfer of lncRNA RP11-838N2. 4 promotes erlotinib resistance in non-small cell lung cancer. Int. J. Oncol. 2018, 53, 527–538.
  87. Kang, M.; Ren, M.; Li, Y.; Fu, Y.; Deng, M.; Li, C. Exosome-mediated transfer of lncRNA PART1 induces gefitinib resistance in esophageal squamous cell carcinoma via functioning as a competing endogenous RNA. J. Exp. Clin. Cancer Res. 2018, 37, 171.
  88. Dong, H.; Wang, W.; Chen, R.; Zhang, Y.; Zou, K.; Ye, M.; He, X.; Zhang, F.; Han, J. Exosome-mediated transfer of lncRNA-SNHG14 promotes trastuzumab chemoresistance in breast cancer. Int. J. Oncol. 2018, 53, 1013–1026.
  89. Lei, Y.; Guo, W.; Chen, B.; Chen, L.; Gong, J.; Li, W. Tumor-released lncRNA H19 promotes gefitinib resistance via packaging into exosomes in non-small cell lung cancer. Oncol. Rep. 2018, 40, 3438–3446.
  90. Cesi, G.; Philippidou, D.; Kozar, I.; Kim, Y.J.; Bernardin, F.; van Niel, G.; Wienecke-Baldacchino, A.; Felten, P.; Letellier, E.; Dengler, S.; et al. A new ALK isoform transported by extracellular vesicles confers drug resistance to melanoma cells. Mol. Cancer 2018, 17, 145.
  91. Xavier, C.P.R.; Castro, I.; Caires, H.R.; Ferreira, D.; Cavadas, B.; Pereira, L.; Santos, L.L.; Oliveira, M.J.; Vasconcelos, M.H. Chitinase 3-like-1 and fibronectin in the cargo of extracellular vesicles shed by human macrophages influence pancreatic cancer cellular response to gemcitabine. Cancer Lett. 2021, 501, 210–223.
  92. Yin, J.; Ge, X.; Shi, Z.; Yu, C.; Lu, C.; Wei, Y.; Zeng, A.; Wang, X.; Yan, W.; Zhang, J.; et al. Extracellular vesicles derived from hypoxic glioma stem-like cells confer temozolomide resistance on glioblastoma by delivering miR-30b-3p. Theranostics 2021, 11, 1763–1779.
  93. Hoshino, A.; Kim, H.S.; Bojmar, L.; Gyan, K.E.; Cioffi, M.; Hernandez, J.; Zambirinis, C.P.; Rodrigues, G.; Molina, H.; Heissel, S.; et al. Extracellular Vesicle and Particle Biomarkers Define Multiple Human Cancers. Cell 2020, 182, 1044–1061.e18.
  94. Kosaka, N.; Yoshioka, Y.; Tominaga, N.; Hagiwara, K.; Katsuda, T.; Ochiya, T. Dark side of the exosome: The role of the exosome in cancer metastasis and targeting the exosome as a strategy for cancer therapy. Future Oncol. 2014, 10, 671–681.
  95. Moloudizargari, M.; Asghari, M.H.; Goel, A. The therapeutic triad of extracellular vesicles: As drug targets, as drugs, and as drug carriers. Biochem. Pharmacol. 2021, 192, 114714.
  96. Santos, M.F.; Rappa, G.; Karbanová, J.; Vanier, C.; Morimoto, C.; Corbeil, D.; Lorico, A. Anti-human CD9 antibody Fab fragment impairs the internalization of extracellular vesicles and the nuclear transfer of their cargo proteins. J. Cell. Mol. Med. 2019, 23, 4408–4421.
  97. Baglio, S.R.; Lagerweij, T.; Pérez-Lanzón, M.; Ho, X.D.; Léveillé, N.; Melo, S.A.; Cleton-Jansen, A.-M.; Jordanova, E.S.; Roncuzzi, L.; Greco, M.; et al. Blocking Tumor-Educated MSC Paracrine Activity Halts Osteosarcoma Progression. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 3721–3733.
More
Information
Subjects: Cell Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 483
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 22 Dec 2021
1000/1000
Video Production Service