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 -- 2099 2022-11-09 13:45:24 |
2 format Meta information modification 2099 2022-11-10 03:24:04 |

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.
Catalano, M.;  Serpe, C.;  Limatola, C. Microglial Extracellular Vesicles for Brain Microenvironment in Glioma. Encyclopedia. Available online: https://encyclopedia.pub/entry/33744 (accessed on 27 July 2024).
Catalano M,  Serpe C,  Limatola C. Microglial Extracellular Vesicles for Brain Microenvironment in Glioma. Encyclopedia. Available at: https://encyclopedia.pub/entry/33744. Accessed July 27, 2024.
Catalano, Myriam, Carmela Serpe, Cristina Limatola. "Microglial Extracellular Vesicles for Brain Microenvironment in Glioma" Encyclopedia, https://encyclopedia.pub/entry/33744 (accessed July 27, 2024).
Catalano, M.,  Serpe, C., & Limatola, C. (2022, November 09). Microglial Extracellular Vesicles for Brain Microenvironment in Glioma. In Encyclopedia. https://encyclopedia.pub/entry/33744
Catalano, Myriam, et al. "Microglial Extracellular Vesicles for Brain Microenvironment in Glioma." Encyclopedia. Web. 09 November, 2022.
Microglial Extracellular Vesicles for Brain Microenvironment in Glioma
Edit

Microglial cells represent the resident immune elements of the central nervous system, where they exert constant monitoring and contribute to preserving neuronal activity and function. In the context of glioblastoma (GBM), a common type of tumor originating in the brain, microglial cells deeply modify their phenotype, lose their homeostatic functions, invade the tumoral mass and support the growth and further invasion of the tumoral cells into the surrounding brain parenchyma. These modifications are, at least in part, induced by bidirectional communication among microglial and tumoral cells through the release of soluble molecules and extracellular vesicles (EVs). EVs produced by GBM and microglial cells transfer different kinds of biological information to receiving cells, deeply modifying their phenotype and activity and could represent important diagnostic markers and therapeutic targets. In GBM, microglial-derived EVs contribute to the immune suppression of the tumor microenvironment (TME), thus favoring GBM immune escape. 

extracellular vesicles microglia glioma

1. Extracellular Vesicles

All living cells are capable of secreting EVs. The production of EVs was initially described as a mechanism for the clearance of unnecessary compounds from the cells [1]. In 1967, researchers identified small lipid vesicles derived from whole serum as well as from platelets after ultracentrifugation. This material was initially referred to as “platelet dust” and later identified as composed of microparticles. In 1971, Aaroson described the membranous structures as EVs [2]. EVs of different sizes were visualized using electron microscopy and it took a long time before the hypothesis that considered EVs as experimental “artifacts” was finally excluded. In 1981, Trams observed that shed “microvesicles” collected from the culture medium of glioblastoma cell lines had a membrane composition similar to specific plasma membrane domains and that these vesicles induced specific effects on receiving cells [3]. Despite the initial evidence, for many years, EVs have been largely overlooked. Only one decade ago, the production of EVs started to be recognized as a new possible mechanism of intercellular communication [4]. EVs have heterogeneous structures delimited by a lipid bilayer and because they lack a functional nucleus, cannot self-replicate. EVs are divided into two categories based on dimension: medium/large EVs (m/lEVs) and small EVs (sEVs). These two populations differ in size, in the mechanism of their formation and in cargo. Concerning the cargo, all EVs can transport molecules with biological activity such as proteins, lipids, and nucleic acids, including DNA and different kinds of RNAs, such as mRNA and microRNA (miRNA). miRNAs are small non-coding molecules able to bind to complementary sequences in the 3′-untranslated regions (3′UTRs) of target mRNAs, defining the post-transcriptional regulation of different genes [5].
Much has been learned on the content of EVs but the identification of cargo specific for small or medium/large vesicles is an ongoing area of intense research.
Because of their capacity to exchange molecules between cells, EVs act as signaling structures both under physiological and pathological conditions. Specifically, in pathological conditions, EVs might act in favor of disease, such as in multiple sclerosis [6], Alzheimer’s [7][8], prion [9], and Huntington’s disease [10]. These aspects, together with the identification of m/lEVs in different body fluids, increased the interest in research to elucidate the vesicle functions in different contexts [11]. In addition, EVs transport biologically active factors across the blood–brain barrier (BBB) and the choroid plexus [12], making them potential tools for the early diagnosis of neurological disease [13] and potential vehicles for non-invasive therapies [14].

1.1. Medium/Large EVs

According to the physical characterization and to the guidelines of the International Society of Extracellular Vesicles (ISEV) [15], m/lEVs comprise all the vesicles with a diameter larger than 200 nm. They are produced by the direct invagination of the plasma membrane and then released into the extracellular space [16].
Each cell type changes the EVs composition according to its physiological state, regardless of its size, with peculiar lipids, proteins, and nucleic acids content [17]. The process of plasma membrane blebbing is typical of m/lEVs, even if the underlying mechanisms remain incompletely understood [17].
Recently, it was reported that the process involved in membrane curvatures during EV formation requires the interaction of the arrestin domain-containing protein-1 (ARRDC1) with the endosomal tumor susceptibility protein 101 (TSG101) and that this interaction leads to the transfer of TSG101 from the endosomal compartment to the plasma membrane. The presence of TSG101 in the plasma membrane allows the release of m/lEVs containing TSG101 and ARRDC1, together with other molecules. Other mechanisms are involved in the process of plasma membrane curvature. For example, the enrichment of proteins at the cellular periphery and the pressure generated by their interaction could contribute to shaping changes and curvature. This process indicates that the enrichment of protein at the site of m/lEVs budding might be a stimulus sufficient to start vesicle formation [18]. Another important factor is the alteration in the lipid composition that modifies membrane rigidity and curvature [19], key events for m/lEV formation. Phospholipids are made up of hydrocarbon tails and large head groups that give them a conical shape, with an irregular distribution. The membrane curvature is determined by the distribution of phospholipids in the plasma membrane but also by the presence of aminophospholipid translocases, such as flippases and floppases, similarly to what is described for the Golgi vesicles [20].

1.2. Small EVs

Differently, sEVs are vesicles with a diameter smaller than 200 nm [15]. At the beginning of the 1980s, a pathway that includes endocytosis was described for sEV formation. It includes the internalization of extracellular ligands and cellular components that will then be transferred again to the plasma membrane and/or degraded [21]. Upon degradation, early endosomes will be the first to be produced, followed by late endosomes [22], which accumulate intraluminal vesicles (ILVs), or multivesicular endosomes (MVBs), inside. Proteins and lipids contained in the MVBs contribute to the curvature of the early endosomal membrane. Mostly, MVBs fuse with lysosomes and their content is degraded by hydrolases. In other cases, they can fuse with clusters of the plasma membrane enriched in tetraspanin CD63, lysosomal-associated membrane proteins (LAMP1 and LAMP2) and other molecules typical of late endosomes, releasing the content into the extracellular space [23][24].
To exert biological effects, EVs can either be internalized or activate receptor–ligand signaling on the surface of target cells. In the last few years, the mechanisms of EVs–cell interaction have also been investigated, taking advantage of drugs or antibodies to block specific signaling pathways. These studies revealed that EVs can be internalized by target cells through different mechanisms, such as clathrin-mediated endocytosis, phagocytosis, micropinocytosis and plasma or endosomal membrane fusion [25][26][27][28][29], in addition to protein–protein interactions mediated by tetraspanins (as CD63, CD9 and CD81), integrins and immunoglobulins, proteoglycan, and lectins [30].

2. Microglial EVs in Brain Tumors

In GBM, microglia and infiltrating macrophages represent about 30% of total cells in the tumor mass, contributing to the early anti-tumor immune response and later playing a role in supporting cancer growth [31][32]. In fact, brain tumor cells attract microglia by secreting factors such as cytokines, growth factors, chemokines and colony-stimulating factors [32], which induce phenotypical and genetical switch of microglia toward a pro-tumorigenic ally. Microglia and infiltrating macrophages, modified by cancer cells, are defined as tumor-associated myeloid cells (TAMCs) [33].
In addition to secreted molecules, microglia-tumor communication is also mediated by gap junctions, tunneling nanotubes and EVs [34]. EVs make possible communication along distant sites and, importantly, also permit bidirectional cell-to-cell communication [34]. As stated before, EVs can deliver not only soluble proteins but also a wide variety of coding and non-coding RNAs that can alter the gene expression of the target cells [34].

3. EV-Mediated Communication between GBM and the Brain Tumor Environment

GBM is the most aggressive brain tumor and represents an important research and medical challenge [35]. EVs released by GBM cells have specific cargos that favor tumor propagation. They contain oncogenes, such as the epidermal growth factor receptor (EGFRvIII), that induce the expression of other oncogenes (i.e., p27 and Bcl-xL) and the activation of pro-tumoral pathways (i.e., AKT and ERK1/2 phosphorylation) [36]. Another mechanism of EV-mediated progression of glioma is the transfer of the RNA-binding motif 11 (RBM11) [37], a pro-tumoral protein that promotes invasion and proliferation [38] by apoptotic glioma cells. Additionally, EVs derived from glioma stem cells (GSCs) contribute to maintaining the GBM cellular heterogeneity (a peculiarity of high malignancy) [39]. In addition to the horizontal propagation of the oncogenic activity, glioma-released EVs also convey materials to non-tumoral cells [40], such as astrocytes, neurons, and endothelial and immune cells.
Astrocytes stimulated by GBM-derived EVs acquire a tumor-supportive phenotype [41]. In addition, GBM-derived EVs enhance the proliferation and the transformation of astrocytes by RNAs able to reprogram metabolic activity [42]. Conversely, exosome-mediated transport of miR-19a from astrocytes to tumor cells critically results in PTEN downregulation and tumor growth [43][44].
GBM-derived EVs can regulate neuronal excitability [45] and miRs transfer (miR-148a and miR-9-5p) promotes angiogenesis in endothelial cells [46][47][48][49]. In bidirectional communication, brain endothelial cells release EVs containing tetraspanin CD9 to GBM [50], enhancing tumor progression through the inhibition of ubiquitination of IL6 receptor gp30 and promoting the activation of the signal transducer and activator of transcription 3 (STAT3) [51][52][53][54]. Endothelial-derived EVs overexpressing the tumor suppressor esophageal cancer-related gene-4 (ECRG4) inhibit glioma cell proliferation [55].
GBM-derived EVs suppress the activation of T lymphocytes, partially through the programmed death ligand-1 (PD-L1) [56], a key player of the tumor immune escape in many cancers including GBM [57][58]. Macrophages stimulated with GBM-derived EVs acquire a pro-tumoral phenotype (i.e., overexpression of Arg-1 and IL-10 and downregulation of iNOS and TNF-α), in part through miR-10b-5p [59]. Furthermore, GBM-released EVs also block the clonal proliferation of T cells by transferring CD73 [60], which inhibits aerobic glycolysis [61].
GBM-derived EVs are involved in the initiation of the tumor-supportive TAMC phenotype [62]. TAMCs efficiently engulf EVs, as visualized in vivo, in glioma-bearing mice [63]. GBM-derived EVs increase the phagocytic activity of microglia towards the extracellular matrix, creating free space in the brain parenchyma [64]. One of the mechanisms involved is the transfer of the membrane type 1-matrix metalloproteinase (MT1-MMP) [64], which degrades the proteins of the extracellular matrix. GBM-derived EVs affect TAMC proliferation both in vivo and in vitro [65]. GBM-derived EVs contain miR-21 [66] which downregulates the target tumor-suppressive gene Btg2 (B cell translocation gene 2) [67] in microglial cells. Moreover, they transfer the Wilms tumor-1 (WT1) protein to microglial cells, upregulating the expression of thrombospondin-1 (Thbs-1), a key promoter of angiogenesis [68]. GSCs represent a cell subpopulation relevant for the tumor resistance to radiations and release EVs able to polarize microglial cells towards a pro-tumoral phenotype, specifically by transferring miR-504 that inhibits the putative onco-suppressor gene grb10 (growth factor receptor-bound protein 10) [66]. miR-504 also increases the pro-tumoral markers CD209 and TGF-β and decreases the expression of the anti-tumoral marker genes CD86 and TNF-α [66]. GBM-released EVs reprogram microglia towards a pro-tumoral phenotype also through the long noncoding RNA (lncRNA) associated with temozolomide (TMZ) (lnc-TALC). lnc-TALC competes with miR-20b-3p by inducing the expression of Stat3, which, in turn, activates the expression of the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT) [69]. lnc-TALC also increases Arg-1, CD163, TGFβ, IL4, and IL10 [70], enhancing the pro-tumoral phenotype of microglia.
On the other side, several pieces of evidence demonstrate that microglial-released EVs can affect the TME and the effects induced by microglial EVs vary with the activation state of these cells [71]. Specifically, m/lEVs released by LPS/INFγ-activated microglia contain transcripts for several inflammation-related genes that reduce the pro-tumoral efficacy of TAMCs. In contrast, m/lEVs released by IL4-stimulated microglia enhance the pro-tumoral phenotype of TAMCs [71]. The biogenesis process and the cargo differ in sEVs and m/lEVs. In fact, in contrast with m/lEVs, sEVs released by both LPS/INFγ and IL4-stimulated microglial cells exert anti-tumoral effects in a mouse model of glioma, reducing tumor mass and prolonging mice survival [72]. Similarly, sEVs released by unstimulated microglial cells reduce the invasion of glioma cells in 3D-spheroid cultures [73]. The anti-glioma effect of microglial-derived sEVs can be mediated by miR124. This specific miR enhances the expression of the glutamate transporter GLT1 on astrocytes, with increased clearance of glutamate in the synaptic cleft. Glioma cells are in fact able to release neurotoxic amounts of glutamate that promote neuronal death and permit the invasion of tumor cells [72].
EV-mediated delivery of miRNAs could represent a promising approach to contrast the growth of brain tumors. In a 3D-microfluidic GBM microenvironment, miR-124-loaded EVs reduce tumor growth and inhibit microglia polarization towards a pro-tumoral phenotype [74]. Furthermore, microglia EVs can be engineered to deliver drugs to glioma cells, taking advantage of their ability to target the brain tumor mass [75]. Specifically, microglia cells were engineered to release EVs enriched in paclitaxel (PTX), a pro-apoptotic compound that blocks the cell cycle at the G2/M phase [76]. Loading PTX in EVs overcomes its low BBB permeability [77] and could be used for other drugs with a reduced capability to enter brain parenchyma.

References

  1. Johnstone, R.M.; Adam, M. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262, 9412–9420.
  2. Aaronson, S.; Behrens, U. Ultrastructure of intracellular and extracellular vesicles, membranes, and myelin figures produced by Ochromonas danica. J. Ultrastruct. Res. 1971, 35, 418–430.
  3. Trams, E.G.; Lauter, C. Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim. Biophys. Acta 1981, 645, 63–70.
  4. Iraci, N.; Gaude, E.; Leonardi, T.; Costa, A.; Cossetti, C.; Peruzzotti-Jametti, L.; Bernstock, J.D.; Saini, H.K.; Gelati, M.; Vescovi, A.L.; et al. Extracellular vesicles are independent metabolic units with asparaginase activity. Nat. Chem. Biol. 2017, 13, 951–955.
  5. de Sousa, M.C.; Gjorgjieva, M.; Dolicka, D.; Sobolewski, C.; Foti, M. Deciphering miRNAs’ Action through miRNA Editing. Int. J. Mol. Sci. 2019, 20, 6249.
  6. Carandini, T.; Colombo, F.; Finardi, A.; Casella, G.; Garzetti, L.; Verderio, C.; Furlan, R. Microvesicles: What is the Role in Multiple Sclerosis? Front. Neurol. 2015, 6, 111.
  7. Aguzzi, A.; Rajendran, L. The Transcellular Spread of Cytosolic Amyloids, Prions, and Prionoids. Neuron 2009, 64, 783–790.
  8. Gouwens, N.W.; Berg, J.; Feng, D.; Sorensen, S.A.; Zeng, H.; Hawrylycz, M.J.; Koch, C.; Arkhipov, A. Systematic generation of biophysically detailed models for diverse cortical neuron types. Nat. Commun. 2018, 9, 710.
  9. Fevrier, B.; Vilette, D.; Archer, F.; Loew, D.; Faigle, W.; Vidal, M.; Laude, H.; Raposo, G. Cells release prions in association with exosomes. Proc. Natl. Acad. Sci. USA 2004, 101, 9683–9688.
  10. Zhang, X.; Abels, E.R.; Redzic, J.S.; Margulis, J.; Finkbeiner, S.; Breakefield, X.O. Potential Transfer of Polyglutamine and CAG-Repeat RNA in Extracellular Vesicles in Huntington’s Disease: Background and Evaluation in Cell Culture. Cell. Mol. Neurobiol. 2016, 36, 459–470.
  11. Fleissner, F.; Goerzig, Y. Microvesicles as novel biomarkers and therapeutic targets in transplantation medicine. Am. J. Transplant. 2012, 12, 289–297.
  12. Balusu, S.; Van Wonterghem, E.; De Rycke, R.; Raemdonck, K.; Stremersch, S.; Gevaert, K.; Brkic, M.; Demeestere, D.; Vanhooren, V.; Hendrix, A.; et al. Identification of a novel mechanism of blood–brain communication during peripheral inflammation via choroid plexus-derived extracellular vesicles. EMBO Mol. Med. 2016, 8, 1162–1183.
  13. Fiandaca, M.S.; Kapogiannis, D.; Mapstone, M.; Boxer, A.; Eitan, E.; Schwartz, J.B.; Abner, E.L.; Petersen, R.C.; Federoff, H.J.; Miller, B.L.; et al. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimer’s Dement. 2015, 11, 600–607.
  14. Alvarez-Erviti, L.; Seow, Y. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345.
  15. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. 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 2018, 7, 1535750.
  16. Tricarico, C.; Clancy, J.; D’Souza-Schorey, C. Biology and biogenesis of shed microvesicles. Small GTPases 2017, 8, 220–232.
  17. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289.
  18. Stachowiak, J.C.; Schmid, E.M.; Ryan, C.J.; Ann, H.S.; Sasaki, D.Y.; Sherman, M.B.; Geissler, P.L.; Fletcher, D.A.; Hayden, C.C. Membrane bending by protein–protein crowding. Nat. Cell Biol. 2012, 14, 944–949.
  19. Stachowiak, J.C.; Brodsky, F.; Miller, E. A cost–benefit analysis of the physical mechanisms of membrane curvature. Nat. Cell Biol. 2013, 15, 1019–1027.
  20. Karaeva, A.R.; Khaskov, M.A.; Mitberg, E.B.; Kulnitskiy, B.A.; Perezhogin, I.A.; Ivanov, L.A.; Denisov, V.N.; Kirichenko, A.N.; Mordkovich, V.Z. Longer Carbon Nanotubes by Controlled Catalytic Growth in the Presence of Water Vapor, Fullerenes, Nanotub. Carbon Nanostruct. 2012, 20, 411–418.
  21. Gould, G.W.; Lippincott-Schwartz, J. New roles for endosomes: From vesicular carriers to multi-purpose platforms. Nat. Rev. Mol. Cell Biol. 2009, 10, 287–292.
  22. Stoorvogel, W.; Strous, G.J.; Geuze, H.J.; Oorschot, V.; Schwartzt, A.L. Late endosomes derive from early endosomes by maturation. Cell 1991, 65, 417–427.
  23. Jaiswal, J.; Andrews, N.; Simon, S.M. Membrane proximal lysosomes are the major vesicles responsible for calcium-dependent exocytosis in nonsecretory cells. J. Cell Biol. 2002, 159, 625–635.
  24. Raposo, G.; Nijman, H.W. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172.
  25. Calvo, V.; Izquierdo, M. Inducible Polarized Secretion of Exosomes in T and B Lymphocytes. Int. J. Mol. Sci. 2020, 21, 2631.
  26. Christianson, H.C.; Svensson, K.J. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their in-ternalization and functional activity. Proc. Natl. Acad. Sci. USA 2013, 110, 173805.
  27. Svensson, K.J.; Christianson, H.C. Exosome uptake depends on ERK1/2-heat shock protein 27 signalling and lipid raft-mediated endocytosis negatively regulated by caveolin-1. J. Biol. Chem. 2013, 288, 1771324.
  28. Morelli, A.E.; Larregina, A.T. Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Blood 2004, 104, 325766.
  29. Tumne, A.; Prasad, V.S. Noncytotoxic suppression of human immunode- ficiency virus type 1 transcription by exosomes se-creted from CD8+ T cells. J. Virol. 2009, 83, 435464.
  30. Thali, M. The Roles of Tetraspanins in HIV-1 Replication. Curr. Top. Microbiol. Immunol. 2009, 339, 85–102.
  31. Borst, K.; Dumas, A.A.; Prinz, M. Microglia: Immune and non-immune functions. Immunity 2021, 54, 2194–2208.
  32. Graeber, M.B.; Scheithauer, B.W.; Kreutzberg, G.W. Microglia in brain tumors. Glia 2002, 40, 252–259.
  33. Hambardzumyan, D.; Gutmann, D.H.; Kettenmann, H. The role of microglia and macrophages in glioma maintenance and progression. Nat. Neurosci. 2016, 19, 20–27.
  34. Skog, J.; Würdinger, T. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 2008, 10, 1470–1476.
  35. Ostrom, Q.T.; Cioffi, G. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2013–2017. Neuro-Oncology 2020, 22, 1–96.
  36. 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.
  37. Pavlyukov, M.S.; Yu, H.; Bastola, S.; Minata, M.; Shender, V.O.; Lee, Y.; Zhang, S.; Wang, J.; Komarova, S.; Wang, J.; et al. Apoptotic Cell-Derived Extracellular Vesicles Promote Malignancy of Glioblastoma Via Intercellular Transfer of Splicing Factors. Cancer Cell 2018, 34, 119–135.e10.
  38. Fu, C.; Yuan, M.; Sun, J.; Liu, G.; Zhao, X.; Chang, W.; Ma, Z. RNA-Binding Motif Protein 11 (RBM11) Serves as a Prognostic Biomarker and Promotes Ovarian Cancer Progression. Dis. Markers 2021, 2021, 3037337.
  39. Ricklefs, F.; Mineo, M.; Rooj, A.K.; Nakano, I.; Charest, A.; Weissleder, R.; Breakefield, X.O.; Chiocca, E.A.; Godlewski, J.; Bronisz, A. Extracellular Vesicles from High-Grade Glioma Exchange Diverse Pro-oncogenic Signals That Maintain Intratumoral Heterogeneity. Cancer Res. 2016, 76, 2876–2881.
  40. Gao, X.; Zhang, Z.; Mashimo, T.; Shen, B.; Nyagilo, J.; Wang, H.; Wang, Y.; Liu, Z.; Mulgaonkar, A.; Hu, X.-L.; et al. Gliomas Interact with Non-glioma Brain Cells via Extracellular Vesicles. Cell Rep. 2020, 30, 2489–2500.e5.
  41. Hallal, S.; Mallawaaratchy, D.M. Extracellular vesicles released by glioblastoma cells stimulate normal astrocytes to acquire a tumor-supportive phenotype via p53 and MYC signaling pathways. Mol. Neurobiol. 2019, 56, 4566–4581.
  42. Zeng, A.; Wei, Z.; Rabinovsky, R.; Jun, H.J.; El Fatimy, R.; Deforzh, E.; Arora, R.; Yao, Y.; Yao, S.; Yan, W.; et al. Glioblastoma-Derived Extracellular Vesicles Facilitate Transformation of Astrocytes via Reprogramming Oncogenic Metabolism. iScience 2020, 23, 101420.
  43. Brandao, M.; Simon, T.; Critchley, G.; Giamas, G. Astrocytes, the rising stars of the glioblastoma microenvironment. Glia 2019, 67, 779–790.
  44. Zhang, L.; Zhang, S.; Yao, J.; Lowery, F.J.; Zhang, Q.; Huang, W.-C.; Li, P.; Li, M.; Wang, X.; Zhang, C.; et al. Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature 2015, 527, 100–104.
  45. Spelat, R.; Jihua, N.; Triviño, C.A.S.; Pifferi, S.; Pozzi, D.; Manzati, M.; Mortal, S.; Schiavo, I.; Spada, F.; Zanchetta, M.E.; et al. The dual action of glioma-derived exosomes on neuronal activity: Synchronization and disruption of synchrony. Cell Death Dis. 2022, 13, 705.
  46. 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.
  47. Kim, J.; Zhang, Y. MicroRNA-148a is a prognostic oncomiR that targets MIG6 and BIM to regulate EGFR and apoptosis in glioblastoma. Cancer Res. 2014, 74, 1541–1553.
  48. Chen, X.; Yang, F. MiR-9 promotes tumorigenesis and angiogenesis and is activated by MYC and OCT4 in human glioma. J. Exp. Clin. Cancer Res. 2019, 38, 99.
  49. Wong, H.A.; Fatimy, R.E. The Cancer Genome Atlas analysis predicts microRNA for targeting cancer growth and vascularization in glioblastoma. Mol. Ther. 2015, 23, 1234–1247.
  50. Podergajs, N.; Motaln, H.; Rajčević, U.; Verbovšek, U.; Koršič, M.; Obad, N.; Espedal, H.; Vittori, M.; Herold-Mende, C.; Miletic, H.; et al. Transmembrane protein CD9 is glioblastoma biomarker, relevant for maintenance of glioblastoma stem cells. Oncotarget 2016, 7, 593–609.
  51. Li, D.; Tian, Y.; Hu, Y.; Qi, Y.; Tian, N.; Li, S.; Hu, P.; Wu, F.; Wei, Q.; Wei, Z.; et al. Glioma-associated human endothelial cell-derived extracellular vesicles specifically promote the tumourigenicity of glioma stem cells via CD9. Oncogene 2019, 38, 6898–6912.
  52. Shi, Y.; Zhou, W. Tetraspanin CD9 stabilizes gp130 by preventing its ubiquitin-dependent lysosomal degradation to promote STAT3 activation in glioma stem cells. Cell Death Differ. 2017, 24, 167–180.
  53. Ouédraogo, Z.G.; Biau, J. Role of STAT3 in genesis and progression of human malignant gliomas. Mol. Neurobiol. 2017, 54, 5780–5797.
  54. Chang, N.; Ahn, S.H. The role of STAT3 in glioblastoma progression through dual influences on tumor cells and the immune microenvironment. Mol. Cell. Endocrinol. 2017, 451, 53–65.
  55. Huo, H.; Yang, S.; Wu, H.; Sun, Y.; Zhao, R.; Ye, R.; Yan, D.; Shi, X.; Yang, J. Brain endothelial cells-derived extracellular vesicles overexpressing ECRG4 inhibit glioma proliferation through suppressing inflammation and angiogenesis. J. Tissue Eng. Regen. Med. 2021, 15, 1162–1171.
  56. 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.
  57. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704.
  58. Wang, H.; Xiao, Y.; Ren, X.; Wan, D. Prognostic value of programmed death ligand 1 (PD-L1) in glioblastoma: A systematic review, meta-analysis and validation based on dataset. Bioengineered 2021, 12, 10366–10378.
  59. Li, B.; Yang, C.; Zhu, Z.; Chen, H.; Qi, B. Hypoxic glioma-derived extracellular vesicles harboring MicroRNA-10b-5p enhance M2 polarization of macrophages to promote the development of glioma. CNS Neurosci. Ther. 2022; epub ahead of print.
  60. Azambuja, J.H.; Gelsleichter, N.E.; Beckenkamp, L.R.; Iser, I.C.; Fernandes, M.C.; Figueiró, F.; Battastini, A.M.O.; Scholl, J.N.; de Oliveira, F.H.; Spanevello, R.M.; et al. CD73 Downregulation Decreases In Vitro and In Vivo Glioblastoma Growth. Mol. Neurobiol. 2019, 56, 3260–3279.
  61. Bavaresco, L.; Bernardi, A.; Braganhol, E.; Cappellari, A.R.; Rockenbach, L.; Farias, P.F.; Wink, M.R.; Delgado-Cañedo, A.; Battastini, A.M.O. The role of ecto-5′-nucleotidase/CD73 in glioma cell line proliferation. Mol. Cell. Biochem. 2008, 319, 61–68.
  62. de Vrij, J.; Maas, S.N.; Kwappenberg, K.M.; Schnoor, R.; Kleijn, A.; Dekker, L.; Luider, T.M.; de Witte, L.D.; Litjens, M.; van Strien, M.E.; et al. Glioblastoma-derived extracellular vesicles modify the phenotype of monocytic cells. Int. J. Cancer 2015, 137, 1630–1642.
  63. Van der Vos, K.E.; Abels, E.R.; Zhang, X.; Lai, C.; Carrizosa, E.; Oakley, D.; Prabhakar, S.; Mardini, O.; Crommentuijn, M.H.W.; Skog, J.; et al. Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro-Oncology 2016, 18, 58–69.
  64. Vinnakota, K.; Hu, F. Toll-like receptor 2 mediates microglia/brain macrophage MT1-MMP expression and glioma expan-sion. Neuro-Oncology 2013, 15, 1457–1468.
  65. Abels, E.R.; Broekman, M.L.; Breakefield, X.O.; Maas, S.L. Glioma EVs Contribute to Immune Privilege in the Brain. Trends Cancer 2019, 5, 393–396.
  66. Bier, A.; Hong, X.; Cazacu, S.; Goldstein, H.; Rand, D.; Xiang, C.; Jiang, W.; Ben-Asher, H.W.; Attia, M.; Brodie, A.; et al. miR-504 modulates the stemness and mesenchymal transition of glioma stem cells and their interaction with microglia via delivery by extracellular vesicles. Cell Death Dis. 2020, 11, 899.
  67. Mao, B.; Zhang, Z.; Wang, G. BTG2: A rising star of tumor suppressors (Review). Int. J. Oncol. 2015, 46, 459–464.
  68. 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.
  69. Wu, P.; Cai, J.; Chen, Q.; Han, B.; Meng, X.; Li, Y.; Li, Z.; Wang, R.; Lin, L.; Duan, C.; et al. Lnc-TALC promotes O6-methylguanine-DNA methyltransferase expression via regulating the c-Met pathway by competitively binding with miR-20b-3p. Nat. Commun. 2019, 10, 2045.
  70. Li, Z.; Meng, X.; Wu, P.; Zha, C.; Han, B.; Li, L.; Sun, N.; Qi, T.; Qin, J.; Zhang, Y.; et al. Glioblastoma Cell–Derived lncRNA-Containing Exosomes Induce Microglia to Produce Complement C5, Promoting Chemotherapy Resistance. Cancer Immunol. Res. 2021, 9, 1383–1399.
  71. Grimaldi, A.; Serpe, C.; Chece, G.; Nigro, V.; Sarra, A.; Ruzicka, B.; Relucenti, M.; Familiari, G.; Ruocco, G.; Pascucci, G.R.; et al. Microglia-Derived Microvesicles Affect Microglia Phenotype in Glioma. Front. Cell. Neurosci. 2019, 13, 41.
  72. Serpe, C.; Monaco, L.; Relucenti, M.; Iovino, L.; Familiari, P.; Scavizzi, F.; Raspa, M.; Familiari, G.; Civiero, L.; D’Agnano, I.; et al. Microglia-Derived Small Extracellular Vesicles Reduce Glioma Growth by Modifying Tumor Cell Metabolism and Enhancing Glutamate Clearance through miR-124. Cells 2021, 10, 2066.
  73. 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.
  74. Hong, S.; You, J.Y.; Paek, K.; Park, J.; Kang, S.J.; Han, E.H.; Choi, N.; Chung, S.; Rhee, W.J.; Kim, J.A. Inhibition of tumor progression and M2 microglial polarization by extracellular vesicle-mediated microRNA-124 in a 3D microfluidic glioblastoma microenvironment. Theranostics 2021, 11, 9687–9704.
  75. Du, Y.; Yang, Z.; Sun, Q.; Lin, M.; Wang, R.; Peng, Y.; Chen, X.; Qi, X. Engineered Microglia Potentiate the Action of Drugs against Glioma Through Extracellular Vesicles and Tunneling Nanotubes. Adv. Health Mater. 2021, 10, e2002200.
  76. Fung, L.K.; Ewend, M.G. Pharmacokinetics of interstitial delivery of carmustine, 4-hydroperoxycyclophosphamide, and paclitaxel from a biodegradable polymer implant in the monkey brain. Cancer Res. 1998, 58, 672–684.
  77. Schinkel, A.H. P-Glycoprotein, a gatekeeper in the blood-brain barrier. Adv. Drug Deliv. Rev. 1999, 36, 179–194.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 300
Revisions: 2 times (View History)
Update Date: 10 Nov 2022
1000/1000
Video Production Service