Skip to main content

OPINION article

Front. Pharmacol., 30 March 2020
Sec. Translational Pharmacology
This article is part of the Research Topic Stem Cells as Targeted Drug Delivery Vehicles View all 10 articles

Cancer Stem Cell-Exosomes, Unexposed Player in Tumorigenicity

  • Stem Cells and Regenerative Medicine Unit, Cell Therapy & Cancer Research Department, King Abdullah International Medical Research Center/King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia

Introduction

Cancer is a well-known, yet poorly understood disease. In which, a healthy tissue is morphed into a cancerous tissue through an intricate, multistep process. This polymorphism has been the focus of cancer research for many decades. Scientists have agreed on a set of traits that are thought to be shared by all cancer tissue types, these traits include; enabling proliferation, evading growth suppressors, resisting cell death, replicative immortality, inducing angiogenesis, and initiating invasion and metastasis, along with other enabling characteristics (Hanahan and Weinberg, 2000; Hanahan and Weinberg, 2011). As researchers investigate the development and propagation of these traits, or as they are called the “hallmarks” of cancer, it became evident that cancer cell-derived extracellular vesicles (EVs), particularly exosomes, play a major role in almost all of them.

In the late 1940s, it was recognized that cells release spherical shaped particles called EVs (Chargaff and West, 1946). Then, almost 40 years later, “exosomes” were acknowledge as a distinct sub-type of EVs (Trams et al., 1981). Up tell now, it is technically challenging to obtain a pure fraction of a specific EV sub-type, due to similarities shared amongst these vesicles. However, the International Society for Extracellular Vesicles, have released a position statement on the minimal experimental requirements for definition of EVs and their functions (MISEV2014; updated in 2018; MISEV2018) (Lotvall et al., 2014; Théry et al., 2018). The MISEV distinction between the different EV sub-types realize on size, density, morphology, subcellular origin, and composition. This was done in order to make scientific reporting on EV biology more consistent and reliable. Most published literature on EVs, including the literature on the role of EVs in cancer, use the term “exosomes” to refer to the EV sub-type under study. These studies include a section that describes the method of “exosome” isolation, and at least a couple of characterization techniques, to justify their nomenclature. Characterization of exosomes in published literature is often based on size and “exosome-enriched” proteins content verification.

On the other hand, the concept of “cancer stem cell” (CSC) only emerged in the 1990s (Lapidot et al., 1994), with a lot of controversy and a number of proposed theories following it. Some say that CSC arise as a result of normal stem cell mutation, while others suggests that CSC arise as a result of a somatic cell acquiring erroneous stem cell characteristics, turning it into cancerous stem cell that can differentiate into heterogeneous population of cancer cells (Baccelli and Trumpp, 2012). Nevertheless, CSCs are now recognized as distinct population of cancer cells, and the CSC-model, is accepted as one of the two most popular models of cancer. The other model being the “clonal evolution-model”, which was described earlier in 1970s. It was postulated that cancer results from the accumulation of mutations in a given group of somatic cell population, within a tissue, thus given raise to heterogeneous population of cancer cells (Nowell, 1976). As the CSC-model becomes more popular, the role of CSCs, as a sub-type of cancer cells, within the tumor microenvironment has recently come to light, especially with advances in stem cell research during the last couple of decades. However, the role of CSC-exosomes, as a sub-type of cancer exosomes, is still under the shadow. Thus, in this article we aim to provide a standpoint on the possible role of CSC-exosomes, and why it should be examined as a separate group of cancer cell-exosomes, based on published literature.

Exosomes, Devoted Messengers for Good or Bad

Exosomes originate from the inward budding of the early endosomes, which later mature into multivesicular bodies (MVBs) (Doyle and Wang, 2019). Depending on their content, MVBs are either sent to the lysosome to be degraded or released into the extracellular space, forming what’s called exosomes (Doyle and Wang, 2019). Cells of different tissue types were found to release exosomes in order to facilitate intercellular communication, thus initiating different biological actions (Ma et al., 2019). Cancer cells, and cancer-associated cells, within the tumor micro-environment were also found to release exosomes. This allows them to commute their message to malignant and non-malignant cells, and initiate pathways that support tumor survival and propagation (Wortzel et al., 2019). The exosome mediated intercellular communication is enabled through “exosomal cargo”. This includes functional proteins, micro-ribonucleic acid (miRNAs) and messenger RNAs (mRNAs) (Hessvik and Llorente, 2018). Exosomes will deliver its cargo from the releasing cell into the recipient cell, which contains the encrypted message. There is a growing body of published literature on the role of cancer cell-exosomes in promoting cancer progression through enabling recipient cells to acquire the mentioned “hall marks” of cancer. A number of studies, have repeatedly shown that cancer cell-exosomes, of different cancer types, significantly increase cancer cell proliferation and inhibit apoptosis by activating various proposed cellular pathways (Zhang et al., 2018; Qian et al., 2019). Studies have also shown that cancer cell-exosomes stimulate angiogenesis by stimulating endothelial cells viability, migration, and tube formation via the transfer of pro‐angiogenic proteins and miRNAs (Yi et al., 2015; Bao et al., 2018; Lin et al., 2018; Yukawa et al., 2018). Likewise, it was reported that cancer cell-exosomes induce replicative immortality via the transfer of telomerase reverse transcriptase mRNA from the telomerase activate cancer cell to the telomerase silenced somatic cell (Gutkin et al., 2016). As for metastasis, it is projected that cancer cells induce metastasis by packing its exosomes with promoters of the epithelial–mesenchymal transition (EMT) cascade, to initiate EMT in the neoplastic epithelial cells, within the tumor microenvironment (Webber et al., 2015; Rahman et al., 2016; Xiao et al., 2016). It is also projected that cancer cells will establish a “pre-metastatic” niche through its exosomes. Cancer cells will release its exosomes into the circulation, where they travel to the metastasis site (Costa-Silva et al., 2015; Liu et al., 2016; Syn et al., 2016). There, cancer cell-exosomes will up-regulate the pro-inflammatory molecules, and vascular leakiness, to mobilize cells that constitute the pre-metastatic niche (Costa-Silva et al., 2015; Liu et al., 2016; Syn et al., 2016). Finally, it is projected that while traveling through the circulation, and engraftment into the new tissue, cancer cell-exosomes support cancer cells by allowing them to escape immune surveillance (Mrizak et al., 2015; Muller et al., 2016; Song et al., 2016). Moreover, in addition to the classical hall marks of cancer, it was reported by a recent study that prostate cancer cell-exosomes play a role in transforming local prostate tissue stem cells into CSCs (Ngalame et al., 2018). While another study reported that glioma cell-exosomes induced a “tumor-like” phenotype in bone-marrow mesenchymal stem cells (BMMSCs) (Ma et al., 2019). This was reported to be based on increased proliferation, migration, and invasion rates of treated BMMSCs. In addition to alteration in BMMSCs protein production, including the production of the metastasis-related proteins.

Cancer Stem Cell, the Black Sheep of the Stem Cell Family

CSCs are cancer cells (found within tumors) that possess characteristics associated with normal stem cells, specifically self-renewal and the ability to differentiate and give rise to different cell types found in a particular cancer specimen i.e. CSCs are tumor-forming cells (Sun et al., 2018). CSCs can be identified by using a set of unified surface markers (i.e. clusters of differentiation (CD); CD44, CD24, CD133), in addition to added tissue specific markers depending on cancer type (Phi et al., 2018). Within the tumor microenvironment, the CSCs are rear and reside in highly specialized niches (Sreepadmanabh and Toley, 2018). The CSCs niche is designed to maintain and protect the CSCs, allowing them to resist many current anticancer treatments (Prieto-Vila et al., 2017). The CSCs niche will also allow the cells to stay dormant for long periods of time, before initiating local recurrent and/or distant metastatic tumors (Plaks et al., 2015). Thus, it is hypothesized that targeting the whole tumor will only slow down tumor expansion while targeting the CSCs, in particular, will jeopardize tumor growth (Garcia-Mayea et al., 2019). At the same time, in regenerative medicine research, it was reported that stem cells and progenitor cells exert their tissue regeneration effects through the release of paracrine factors, mainly exosomes. Studies are consistently showing that injecting the cell-derived exosomes alone, is enough to induce the same regenerative effect as the “whole-cell” transplant approach. For example, it was reported that exosomes derived from embryonic stem cells (Khan et al., 2015), BMMSCs (Zou et al., 2019), and cardiac progenitor cells (Kervadec et al., 2016), all mimic the benefits of injecting their parent cells in a chronic heart failure and myocardial infarction animal models. Thus, it is logical to assume that CSCs function through the same mechanism as other cancer cells and non-cancer stem cells. We can project that CSCs fulfill its “stemness duties” through the release of paracrine factors, with exosomes as a key player.

What Is Proposed?

As discussed above, cancer cell-exosomes are crucial for tumor initiation, maintenance, and propagation. However, published literature on this subject matter often don’t describe the sub-type of cancer cells that these exosomes were derived from. It is well established by now that cancer cell-exosomes mediate cell to cell communication within the tumor microenvironment, to support and promote tumorigenesis. It is also well established by now that any alteration to parent cell, alters exosome secretion and content, which in turn alters its message. For example, when cancer cells were subjected to hypoxia prior to exosome isolation, to reflect the tumor’s hypoxic environment, these exosomes significantly increased migration and invasion of cancer cells (Li et al., 2016), and tube formation by endothelia cells (Kucharzewska et al., 2013; Hsu et al., 2017), compared with exosomes derived from normoxic cancer cells. Therefore, it could be hypothesized that the sub-population of cancer cells, CSCs, produce exosomes with unique characteristics, and thus functions. Currently, there are only few reports on “CSC-derived exosomes”, and their role in cancer propagation, compared to “non-stem cancer cell-derived exosomes” (Table 1). One of the first studies to address this issue reported that the “macrovesicles” that had the in vitro and in vivo angiogenic effect, in renal cancer, were those driven from the CD105+ cancer cell sub-population (Grange et al., 2011). Then later on, one study did a miRNA content comparison, and reported that prostate CSC-derived exosomes have in fact a different miRNA content compared with non-stem prostate cancer cell-derived exosomes (Sánchez et al., 2016). Then, a following study reported that glioma stem cell-derived exosomes promoted angiogenesis by containing a particularly high levels of miRNA-21, which upregulates the vascular endothelial growth factor (VEGF) (Sun et al., 2017). While another study identified 11 miRNAs that are characteristic of gastric CSC-derived exosomes, and suggested that a measurement of these miRNAs in patient serum could be used as a predictor of cancer metastasis (Sun et al., 2017). Other recent CSC-exosomes investigations focusing on their role in metastasis, reported that CSC-derived exosomes promote metastasis by promoting EMT in renal cell carcinoma (Wang et al., 2019) and thyroid cancer (Hardin et al., 2018) via the transfer of miRNA-19b-3p and non-coding-RNAs respectively. Whereas other reported on CSC-exosome role in creating a pro-tumoral microenvironment. For example, it was reported that glioblastoma stem cell-derived exosomes direct monocytes toward the immune suppressive “M2” phenotype, through the signal transducer and activator of transcription-3 (STAT3) pathway, creating an immunosuppressive microenvironment (Gabrusiewicz et al., 2018). While colorectal cancer stem cell-derived exosomes promote a pro-tumoral phenotype in neutrophils by increasing interleukin-(IL)-1β expression (Hwang et al., 2019)

TABLE 1
www.frontiersin.org

Table 1 Summary of published work on the distinct role of CSC-derived exosomes in tumorigenicity.

Since tumor–host cross-talk is believed to be initiated by CSCs, and communication between cancer cells and other cells is conducted through exosomes, it’s of great importance to take a closer look at the role of CSCs-exosomes, and its involvement in tumor aggressiveness. Also, to examine their miRNA content, compared to non-stem cancer cell- exosomes, in order to postulate mechanisms of actions. Then finally, develop a cancer management strategy that targets CSCs, and involves blockage of the CSC-exosome release channels.

Discussion

CSCs generate tumors through the stem cell processes of self-renewal and differentiation into multiple malignant cell types. Based on advances in cell signaling biology, it’s expected that these CSCs function through its exosomes. The term “exosome” was used in this article due to the fact that published literature describing EVs role in cancer often refer to the EV sub-type being examined as exosomes. These publications offer reasonable evidence that the EV sub-type being examined is in fact exosomes, via various methods of characterization. Other sub-types of EVs i.e. ectosomes, microvesicle particles, and apoptotic bodies, could be released by cancer cells/CSCs, and could play a role as well. However there is no adequate reporting on this in the literature. Therefore, based on findings on the role of cancer cell- exosomes, and the role of CSCs in cancer, the role of “CSC-exosomes” should be investigated as a separate entity. Such studies will encounter a significant technical and quality control issues related to harvestation of a pure CSC population, and subsequent yield of pure CSC-exosome fraction. Nevertheless, the knowledge provided by these studies will be crucial in developing a more effective approaches to control progression and metastasis of tumors and prevent recurrence.

Author Contributions

BA-S conceptualized and wrote the article. Other authors were involved in manuscript review and editing.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

EV, Extracellular Vesicle; CSC, Cancer Stem Cell; MVBs, Multivesicular Bodies; miRNA, micro Ribonucleic Acid; mRNA, messenger Ribonucleic Acid; EMT, Epithelial–Mesenchymal Transition; BMMSC, Bone-Marrow Mesenchymal Stem Cell; CD, Clusters of Differentiation; VEGF, Vascular Endothelial Growth Factor; STAT3; Signal Transducer and Activator of Transcription-3; IL, Interleukin.

References

Baccelli, I., Trumpp, A. (2012). The evolving concept of cancer and metastasis stem cells. J. Cell Biol. 198 (3), 281–293. doi: 10.1083/jcb.201202014

PubMed Abstract | CrossRef Full Text | Google Scholar

Bao, L., You, B. O., Shi, S. I., Shan, Y., Zhang, Q., Yue, H., et al. (2018). Metastasis-associated miR-23a from nasopharyngeal carcinoma-derived exosomes mediates angiogenesis by repressing a novel target gene TSGA10. Oncogene 37 (21), 2873–2889. doi: 10.1038/s41388-018-0183-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Chargaff, E., West, R. (1946). The biological significance of the thromboplastic protein of blood. J. Biol. Chem. 166 (1), 189–197.

PubMed Abstract | Google Scholar

Costa-Silva, B., Aiello, N. M., Ocean, A. J., Singh, S., Zhang, H., Thakur, B. K., et al. (2015). Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 17 (6), 816–826. doi: 10.1038/ncb3169

PubMed Abstract | CrossRef Full Text | Google Scholar

Doyle, L. M., Wang, M. Z. (2019). Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 8 (7), 727. doi: 10.3390/cells8070727

CrossRef Full Text | Google Scholar

Gabrusiewicz, K., Li, X., Wei, J., Hashimoto, Y., Marisetty, A. L., Ott, M., et al. (2018). Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology 7 (4), e1412909. doi: 10.1080/2162402X.2017.1412909

PubMed Abstract | CrossRef Full Text | Google Scholar

Garcia-Mayea, Y., Aiello, N. M., Ocean, A. J., Singh, S., Zhang, H., Thakur, B. K., et al. (2019). Insights into new mechanisms and models of cancer stem cell multidrug resistance. Semin. Cancer Biol.

Google Scholar

Grange, C., Tapparo, M., Collino, F., Vitillo, L., Damasco, C., Deregibus, M. C., et al. (2011). Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 71 (15), 5346–5356. doi: 10.1158/0008-5472.CAN-11-0241

PubMed Abstract | CrossRef Full Text | Google Scholar

Gutkin, A., Uziel, O., Beery, E., Nordenberg, J., Pinchasi, M., Goldvaser, H., et al. (2016). Tumor cells derived exosomes contain hTERT mRNA and transform nonmalignant fibroblasts into telomerase positive cells. Oncotarget 7 (37), 59173–59188. doi: 10.18632/oncotarget.10384

PubMed Abstract | CrossRef Full Text | Google Scholar

Hanahan, D., Weinberg, R. A. (2000). The hallmarks of cancer. Cell 100 (1), 57–70. doi: 10.1016/S0092-8674(00)81683-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Hanahan, D., Weinberg, R. A. (2011). Hallmarks of cancer: the next generation. Cell 144 (5), 646–674. doi: 10.1016/j.cell.2011.02.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Hardin, H., Helein, H., Meyer, K., Robertson, S., Zhang, R., Zhong, W., et al. (2018). Thyroid cancer stem-like cell exosomes: regulation of EMT via transfer of lncRNAs. Lab. Invest. 98 (9), 1133–1142. doi: 10.1038/s41374-018-0065-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Hessvik, N. P., Llorente, A. (2018). Current knowledge on exosome biogenesis and release. Cell Mol. Life Sci. 75 (2), 193–208. doi: 10.1007/s00018-017-2595-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Hsu, Y. L., Hung, J. Y., Chang, W. A., Lin, Y. S., Pan, Y. C., Tsai, P. H., et al. (2017). Hypoxic lung cancer-secreted exosomal miR-23a increased angiogenesis and vascular permeability by targeting prolyl hydroxylase and tight junction protein ZO-1. Oncogene 36 (34), 4929–4942. doi: 10.1038/onc.2017.105

PubMed Abstract | CrossRef Full Text | Google Scholar

Hwang, W. L., Lan, H. Y., Cheng, W. C., Huang, S. C., Yang, M. H. (2019). Tumor stem-like cell-derived exosomal RNAs prime neutrophils for facilitating tumorigenesis of colon cancer. J. Hematol. Oncol. 12 (1), 10. doi: 10.1186/s13045-019-0699-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Kervadec, A., Bellamy, V, El Harane, N., Arakéan, L., Vanneaux, V., Cacciapuoti, I., et al. (2016). Cardiovascular progenitor-derived extracellular vesicles recapitulate the beneficial effects of their parent cells in the treatment of chronic heart failure. J. Heart Lung Transplant. 35 (6), 795–807. doi: 10.1016/j.healun.2016.01.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Khan, M., Nickoloff, E., Abramova, T., Johnson, J., Verma, S. K., Krishnamurthy, P., et al. (2015). Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ. Res. 117 (1), 52–64. doi: 10.1161/CIRCRESAHA.117.305990

PubMed Abstract | CrossRef Full Text | Google Scholar

Kucharzewska, P., Christianson, H. C., Welch, J. E., Svensson, K. J., Fredlund, E., Ringn, M., et al. (2013). Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc. Natl. Acad. Sci. U S A 110 (18), 7312–7317. doi: 10.1073/pnas.1220998110

PubMed Abstract | CrossRef Full Text | Google Scholar

Lapidot, T., Sirard, C., Vormoor, J., Murdoch, B., Hoang, T., Caceres-Cortes, J., et al. (1994). A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367 (6464), 645–648. doi: 10.1038/367645a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, L., Li, C., Wang, S., Wang, Z., Jiang, J., Wang, W., et al. (2016). Exosomes Derived from Hypoxic Oral Squamous Cell Carcinoma Cells Deliver miR-21 to Normoxic Cells to Elicit a Prometastatic Phenotype. Cancer Res. 76 (7), 1770–1780. doi: 10.1158/0008-5472.CAN-15-1625

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, X. J., Fang, J. H., Yang, X. J., Zhang, C., Yuan, Y., Zheng, L., et al. (2018). Hepatocellular Carcinoma Cell-Secreted Exosomal MicroRNA-210 Promotes Angiogenesis In Vitro and In Vivo. Mol. Ther. Nucleic Acids 11, 243–252. doi: 10.1016/j.omtn.2018.02.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Gu, Y., Han, Y., Zhang, Q., Jiang, Z., Zhang, X., et al. (2016). Tumor Exosomal RNAs Promote Lung Pre-metastatic Niche Formation by Activating Alveolar Epithelial TLR3 to Recruit Neutrophils. Cancer Cell 30 (2), 243–256. doi: 10.1016/j.ccell.2016.06.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Lötvall, J., Hill, A. F., Hochberg, F. (2014). Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J. Extracell Vesicles 3, 26913. doi: 10.3402/jev.v3.26913

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, Z., Cui, X., Lu, L., Chen, G., Yang, Y., Hu, Y., et al. (2019). Exosomes from glioma cells induce a tumor-like phenotype in mesenchymal stem cells by activating glycolysis. Stem Cell Res. Ther. 10 (1), 60. doi: 10.1186/s13287-019-1149-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Mrizak, D., Martin, N., Barjon, C., Jimenez-Pailhes, A. S., Mustapha, R., Niki, T., et al. (2015). Effect of nasopharyngeal carcinoma-derived exosomes on human regulatory T cells. J. Natl. Cancer Inst 107 (1), 363. doi: 10.1093/jnci/dju363

PubMed Abstract | CrossRef Full Text | Google Scholar

Muller, L., Mitsuhashi, M., Simms, P., William, E. G., Whiteside, T. L. (2016). Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Sci. Rep. 6, 20254. doi: 10.1038/srep20254

PubMed Abstract | CrossRef Full Text | Google Scholar

Ngalame, N. N., Luz, A.L., Makia, N., Tokar, E. J. (2018). Arsenic Alters Exosome Quantity and Cargo to Mediate Stem Cell Recruitment Into a Cancer Stem Cell-Like Phenotype. Toxicol. Sci. 165 (1), 40–49. doi: 10.1093/toxsci/kfy176

PubMed Abstract | CrossRef Full Text | Google Scholar

Nowell, P. C. (1976). The clonal evolution of tumor cell populations. Science 194 (4260), 23–28. doi: 10.1126/science.959840

PubMed Abstract | CrossRef Full Text | Google Scholar

Phi, L. T., Sari, I. N., Yang, Y. G., Lee, S. H., Jun, N., Kim, K. S., et al. (2018). Cancer Stem Cells (CSCs) in Drug Resistance and their Therapeutic Implications in Cancer Treatment. Stem Cells Int. 2018, 5416923. doi: 10.1155/2018/5416923

PubMed Abstract | CrossRef Full Text | Google Scholar

Plaks, V., Kong, N., Werb, Z. (2015). The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 16 (3), 225–238. doi: 10.1016/j.stem.2015.02.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Prieto-Vila, M., Takahashi, R. U., Usuba, W., Kohama, I., Ochiya, T. (2017). Drug Resistance Driven by Cancer Stem Cells and Their Niche. Int. J. Mol. Sci. 18 (12), 2574. doi: 10.3390/ijms18122574

CrossRef Full Text | Google Scholar

Qian, S., Tan, X., Liu, X., Liu, P., Wu, Y. (2019). Exosomal Tenascin-c induces proliferation and invasion of pancreatic cancer cells by WNT signaling. Onco Targets Ther. 12, 3197–3205. doi: 10.2147/OTT.S192218

PubMed Abstract | CrossRef Full Text | Google Scholar

Rahman, M. A., Barger, J. F., Lovat, F., Gao, M., Otterson, G. A., Nana-Sinkam, P., et al. (2016). Lung cancer exosomes as drivers of epithelial mesenchymal transition. Oncotarget 7 (34), 54852–54866. doi: 10.18632/oncotarget.10243

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanchez, C. A., Andahur, E. I., Valenzuela, R., Castellón, E. A., Fullá, J. A., Ramos, C. G., et al. (2016). Exosomes from bulk and stem cells from human prostate cancer have a differential microRNA content that contributes cooperatively over local and pre-metastatic niche. Oncotarget 7 (4), 3993–4008. doi: 10.18632/oncotarget.6540

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, X., Ding, Y., Liu, G., Yang, X., Zhao, R., Zhang, Y., et al. (2016). Cancer Cell-derived Exosomes Induce Mitogen-activated Protein Kinase-dependent Monocyte Survival by Transport of Functional Receptor Tyrosine Kinases. J. Biol. Chem. 291 (16), 8453–8464. doi: 10.1074/jbc.M116.716316

PubMed Abstract | CrossRef Full Text | Google Scholar

Sreepadmanabh, M., Toley, B. J. (2018). Investigations into the cancer stem cell niche using in-vitro 3-D tumor models and microfluidics. Biotechnol. Adv. 36 (4), 1094–1110. doi: 10.1016/j.biotechadv.2018.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, X., Ma, X., Wang, J., Zhao, Y., Wang, Y., Bihl, J. C., et al. (2017). Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal. Oncotarget 8 (22), 36137–36148. doi: 10.18632/oncotarget.16661

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, Z. P., Li, A. Q., Jia, W. H., Ye, S., Van Eps, G., Yu, J. M., et al. (2017). MicroRNA expression profiling in exosomes derived from gastric cancer stem-like cells. Oncotarget 8 (55), 93839–93855. doi: 10.18632/oncotarget.21288

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, Z., Wang, L., Dong, L., Wang, X. (2018). Emerging role of exosome signalling in maintaining cancer stem cell dynamic equilibrium. J. Cell Mol. Med. 22 (8), 3719–3728. doi: 10.1111/jcmm.13676

CrossRef Full Text | Google Scholar

Syn, N., Wang, L., Sethi, G., Thiery, J. P., Goh, B. C. (2016). Exosome-Mediated Metastasis: From Epithelial-Mesenchymal Transition to Escape from Immunosurveillance. Trends Pharmacol. Sci. 37 (7), 606–617. doi: 10.1016/j.tips.2016.04.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Thery, C., Witwer, K. W., Aikawa, E., Alcaraz, M. J., Anderson, J. D., Andriantsitohaina, R., et al. (2018). Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell Vesicles 7 (1), 1535750. doi: 10.1080/20013078.2018.1535750

PubMed Abstract | CrossRef Full Text | Google Scholar

Trams, E. G., Lauter, C. J., Salem, J. N., Heine, U. (1981). Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim. Biophys. Acta 645 (1), 63–70. doi: 10.1016/0005-2736(81)90512-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., Yang, G., Zhao, D., Wang, J., Bai, Y., Peng, Q., et al. (2019). CD103-positive CSC exosome promotes EMT of clear cell renal cell carcinoma: role of remote MiR-19b-3p. Mol. Cancer 18 (1), 86. doi: 10.1186/s12943-019-0997-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Webber, J. P., Spary, L. K., Sanders, A. J., Chowdhury, R., Jiang, W. G., Steadman, R., et al. (2015). Differentiation of tumour-promoting stromal myofibroblasts by cancer exosomes. Oncogene 34 (3), 290–302. doi: 10.1038/onc.2013.560

PubMed Abstract | CrossRef Full Text | Google Scholar

Wortzel, I., Dror, S., Kenific, C. M., Lyden, D. (2019). Exosome-Mediated Metastasis: Communication from a Distance. Dev. Cell 49 (3), 347–360. doi: 10.1016/j.devcel.2019.04.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, D., Barry, S., Kmetz, D., Egger, M., Pan, J., Rai, S. N., et al. (2016). Melanoma cell-derived exosomes promote epithelial-mesenchymal transition in primary melanocytes through paracrine/autocrine signaling in the tumor microenvironment. Cancer Lett. 376 (2), 318–327. doi: 10.1016/j.canlet.2016.03.050

PubMed Abstract | CrossRef Full Text | Google Scholar

Yi, H., Ye, J., Yang, X. M., Zhang, L. W., Zhang, Z. G., Chen, Y. P. (2015). High-grade ovarian cancer secreting effective exosomes in tumor angiogenesis. Int. J. Clin. Exp. Pathol. 8 (5), 5062–5070.

PubMed Abstract | Google Scholar

Yukawa, H., Suzuki, K., Aoki, K., Arimoto, T., Yasui, T., Kaji, N., et al. (2018). Imaging of angiogenesis of human umbilical vein endothelial cells by uptake of exosomes secreted from hepatocellular carcinoma cells. Sci. Rep. 8 (1), 6765. doi: 10.1038/s41598-018-24563-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, P., Zhou, H., Lu, K., Lu, Y., Wang, Y., Feng, T. (2018). Exosome-mediated delivery of MALAT1 induces cell proliferation in breast cancer. Onco Targets Ther. 11, 291–299. doi: 10.2147/OTT.S155134

PubMed Abstract | CrossRef Full Text | Google Scholar

Zou, L., Ma, X., Lin, S., Wu, B., Chen, Y., Peng, C. (2019). Bone marrow mesenchymal stem cell-derived exosomes protect against myocardial infarction by promoting autophagy. Exp. Ther. Med. 18 (4), 2574–2582. doi: 10.3892/etm.2019.7874

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: cancer stem cell (CSC), exosome, cancer, cancer cell-derived exosomes, stem cell

Citation: Al-Sowayan BS, Al-Shareeda AT and Alrfaei BM (2020) Cancer Stem Cell-Exosomes, Unexposed Player in Tumorigenicity. Front. Pharmacol. 11:384. doi: 10.3389/fphar.2020.00384

Received: 21 November 2019; Accepted: 12 March 2020;
Published: 30 March 2020.

Edited by:

Rebecca Lim, Hudson Institute of Medical Research, Australia

Reviewed by:

Sonia A. Melo, University of Porto, Portugal
Zui Pan, University of Texas at Arlington, United States

Copyright © 2020 Al-Sowayan, Al-Shareeda and Alrfaei. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Batla S. Al-Sowayan, alsowayanba@ngha.med.sa

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.