Abstract
Seminal discoveries have established the role of complex tumor microenvironment (TME) in cancer progression; and later on also uncovered that vesiculation is an integral part of intercellular communication among various cell types in coordinating the tumor assembly in a dynamic manner. Exosomes are small membrane bound endosomal vesicles, which are classically known for their role in discarding cellular wastes; however, recent reports underlined their novel role in malignancy by their release from cells into the TME. Since then, the role of exosomes have been a subject of increasing interest, as exosome mediated intercellular communications offer a novel reciprocal relationship between cancer and stromal cells within the TME and modulate the fate and function of the recipient cells to finally shape the tumor progression. Exosomes are characterised by different features including size, content and mode of delivery; and its cargo delivers interesting bioactive components in the form of proteins, miRNAs or other molecules to the target cell. In the pursuit of further study of exosomes, it was found that with the help of its distinct bioactive components, exosomes specifically regulate tumor growth, angiogenesis, metastasis as well as drug resistance properties. In fact, it acts as a bridge between different signaling networks, present inside the spatially distant cells of the heterogeneous tumor population. In the current endeavour, we have highlighted the role of exosomes in modulating the intercellular crosstalk during tumor growth and progression, and proposed certain novel roles of exosomes to address the few enigmatic questions of cancer cell biology.
Keywords: Exosome, Tumor Microenvironment, Angiogenesis, Metastasis, Drug Resistance
1. Introduction
Cell secretion is a widely accepted phenomenon and a fundamental process for the maintenance of physiological functions of a cell1. It is accompanied mostly by microvesicles or exosome like vesicles which were previously known by many names depending on their cellular origin: ectosome, oncosome, texosome, prostasome, epididymosome and dexosome2,3. The concept of extracellular vesicles (EV) being introduced, in 1980s by Johnstone’s and Stahl’s groups, for secretory vesicles originating from endosome in multivesicular bodies with a function of clearing transferrin receptor during reticulocyte maturation. However, the word ‘exosome’ was proposed for the first time for EV of endosomal origin in 19874-6. Exosomes are small lipid bilayer vesicles secreted by most but not all types of cells in their microenvironment. Besides their well known role in discarding waste material from the cells, exosomes play an important function in maintaining normal as well as pathological processes7. In case of tumor development and progression, exosome has emerged as an important mediator of cellular communication and opened up a window that extends our understanding about how certain secretory vesicles perform a critical function of transferring genetic material, induce epigenetic changes, modulate immune response to manipulate the local and systemic tumor environment to regulate cancer growth and dissemination. Current endeavour is an attempt to shed light on the emerging role of tumor-secreted exosomes as a novel player to modulate the tumor microenvironment during carcinogenesis.
2. Exosomes: Isolation and Characterization
Cells secrete different kinds of vesicles in the body fluids, which vary in their biogenesis as well as in biophysical properties. Exosomes have certain characteristic features including their size (40 to 100 nm) and morphology that distinguish them from other EVs8. Though the role of exosomes in regulating the fate of other cells within the tissue is now widely accepted, controversies exist for the terminology in the field of EV research and they are sometimes misleading per se in the context of their functionality. As an attempt to address such controversies, releasing mechanism of EVs in the extracellular environment has been used as the criteria for its nomenclature. On this basis, EVs can be categorized into (i) exosomes: 40-100 nm diameter membranous vesicles of endocytic origin released by exocytic fusion of multivesicular bodies to plasma membrane, (ii) ectosomes (also referred to as shedding microvesicles): large membranous vesicles of 50-1000 nm diameter that are shed directly from the plasma membrane, and (iii) apoptotic blebs (50-5000 nm diameter): released by dying cells9-13. Similar to other vesicles, exosomes contain integrins, selectins but unlike others, they also harbour annexins, Rab proteins, SNAREs (v-SNARE and t-SNARE), tetraspanins like CD9, CD82, CD81, CD63 etc. and enrichment in lipids like cholesterol, sphingolipids, ceramides and glycerophospholipids with long saturated fatty acyl chains8,14-17.
Recent studies have shown that exosomes can be isolated in vivo from body fluids such as blood, urine, breast milk, amniotic fluid, malignant ascites, bronchoalveolar lavage fluid and synovial fluid etc18-25. These vesicles can be isolated by various techniques including differential ultracentrifugation, which can be sometimes combined with 0.1 μm to 0.22 μm filtration in order to separate the nano-sized particles from larger particles and cellular debris26. Immuno-affinity technique using beads against tetraspanins, CD63 or CD82, or other origin specific exosomal markers is also used to isolate these vesicles27. Unfortunately, the purification methods are often confusing in distinguishing exosomes from other EVs present as cross contaminations9. Microscopy based estimation of exosomes has its own limitation of fixation and dehydration, that may result in shrinkage and hence underestimation of actual size. However, certain modern techniques like nanoparticle based tracking analysis can be used to overcome these problems by analyzing them in PBS based on their Brownian motion in suspension to avoid shrinkage28,29. As conventional flow cytometers cannot differentiate between vesicles that are less than 300 nm, a novel high resolution flow cytometry–based method has been discovered for a quantitative high throughput analysis of individual (immuno-labelled) nano-sized vesicles30-32.
Cargo material of an exosome may include lipids, proteins (oncoproteins, tumor suppressor proteins and functional transmembrane proteins), nucleic acids (mRNA, miRNA, DNA), growth receptors, and soluble factors33-40. Various techniques including western blot, mass spectrometry, fluorescence activated cell sorting and immuno-electron microscopy can be used to analyse the exosomal content41. At a given time point, exosomes do not represent the snapshot of transcriptome of parental cells but exhibit a selective cargo profiles42-44. For example, exosomes from Glioma represent a different repertoire of microRNAs compared to their cells of origin, with abundance of unusual or novel non-coding RNAs of unknown functions45. Furthermore, it has been found that there is a low correlation between exosomal miRNA content of metastatic SKOV-3 cells versus non-metastatic OVCAR-3 cells. This dictates that specific export of miRNAs may have some potential tumor-specific modulatory role46.
3. Exosomes: Release and Uptake
It is well established that exosomes are released by the cells after fusion of multivesicular bodies with the plasma membrane, but the molecular mechanism of their release to the microenvironment is still poorly understood47. It has been found that a number of Rab family proteins, such as Rab22a, Rab27a, Rab27b and Rab35, R-SNARE protein YKT6, glyco-sphingolipids and flotillins, as well as ceramides, are involved in the regulation of their secretion17,48-52. Nevertheless, the involvement of these proteins in exosome formation is dependent on cell types. Exosomal release has been found to be dependent on several physico-chemical factors like calcium, hypoxia, chemotherapeutic drug exposure, temperature and oxidative stress etc44,53-55 Recently, a group of scientists explored a novel feedback mechanism to control the exosomal release in a cell type specific manner. They found that exosomes from normal mammary epithelial cells at a similar concentration had a dramatic inhibitory effect on exosome production by breast cancer cells compared to exosomes from bladder cancer cells. They also proposed a dynamic equilibrium between the release and uptake of exosomes from and to the surrounding medium, depending on their concentration in extracellular environment (Figure 1)56.
Figure 1. Schematic representation of exosome mediated intercellular crosstalk in tumor microenvironment.
The exosomal uptake is carried out by either endocytosis or phagocytosis or micro-pinocytosis57-59.In 2009, Parolini et al. demonstrated another way of exosome import to receiving cells by lipid dependant membrane fusion through "lipid rafts" of exosomal membrane. They also suggested that low pH microenvironment increases the uptake of exosomes as well as their release from the cancer cells, probably because of the intrinsically endowed negative charge due to high lipid content in exosomes. This could be the possible reason behind the cancer cells to secrete higher amount of exosomes compared to their normal counterpart and their preferential delivery to metastatic tumor cells rather than primary tumor site60.
4. Exosomes and Tumor Microenvironment (TME)
TME is composed of various cells which include stromal cells like endothelial cells, carcinoma-associated fibroblasts (CAFs), adipocytes, mesenchymal cells, immune cells (tumor-associated macrophages or TAM, T-cells etc.), as well as extracellular matrix which encircle all tumor cells, and soluble factors such as growth factors, chemokines and cytokines61. Besides genetic and epigenetic control, cells of the TME also possess a remodelling influence on the tumor growth and its transformation from benign to malignant. For tumor propagation, cellular communication is not only required between the tumor cells but also between cancer cells and the other neighbouring cells in TME. However, such intercellular communication can occur with or without cell-to-cell contact. Contact independent communication may take place via endocrine signaling or as an alternative; exosomes represent another way of distant cellular communication. This exosomal cross talk between resident cells imparts strong influence on inherent complexity of TME by regulating and moulding various oncogenic signaling pathways.
4.1 Role in Tumor Growth
In a normal cellular homeostatic condition, cells maintain a competitive environment for deleterious cells to eradicate them from the local niche but during tumor initiation, transformed cells dominate over normal cells. Recent experimental evidence suggest an important role of exosome mediated intercellular communication in this tug of war of normal versus tumorigenic cells (Figure 1)62. As for example, exosomal miR-143 derived from non-cancerous cells have the ability to suppress the growth of cancer cells both in vitro and in vivo63. In contrast, breast cancer cell (MDA-MB-231, T47D:A18 and MCF-7) derived exosomes manipulate epithelial cells of the mammary duct to facilitate tumor development64.
During tumor growth, Wnt signaling play a pivotal role where Wnt ligands disperse spatially, however, the mechanism of their dispersal remained enigmatic. Recently, exosome mediated transfer of proteins resolve the riddle of how the hydrophobic Wnts traffic over long distances. In this context, Wnt3A has been found to be present on the surface of exosomes which explains a putative mechanism of Wnt transport to the distant recipient cells.51 Tumor suppressor Phosphatase and tensin homolog (PTEN) is a well established intracellular signaling molecule, but, interestingly, it can also be transported outside the cells via exosomal cargo and impart growth inhibitory function35,65.
Tumor derived exosomes can also influence the immune system by triggering the immunosuppressive responses, in order to favor tumor progression. These exosomes can stimulate the expansion of regulatory T-cells, which in turn impair the function of anti-tumorigenic T-cells in TME66. It has also been found that Transforming growth factor-beta (TGF-β) present on the surface of exosomes influences the immunosuppressive effects of regulatory T-cells67. Tumor derived microvesicles (MVs) also carry Fas ligands and TRAIL on their surface, which indicates their apoptosis inducing effect on activated T-cells68,69. Moreover, enzymes of exosomes negatively regulate T-cell activation by hydrolysis of ATP into adenosine70. Secretory exosomes also reduce the proliferation of Natural Killer (NK) cells or block the Interleukin 2-mediated activation of NK cells71,72.
4.2 Role in Angiogenesis
The process of angiogenesis requires a balance between pro- and anti-angiogenic factors in order to disseminate the endothelial progenitor cells to the vasculogenic site. Among the various factors, vascular endothelial growth factor (VEGF) and their receptors play a crucial role in maintaining the vascular homeostasis. During tumorigenesis, TAMs secrete VEGF, which in turn leads to vascularization to sustain tumor growth. Interestingly, microvesicle- delivery of antisense miR-150 into the mice decreased the secretion of VEGF by TAMs via targeting ING473. Although the expression of EGFR on endothelial cells has been controversial, recently it has been found that exosomes facilitate a path for the expression of EGFR on the HUVEC as well as tumor associated endothelial cells, through phosphotidylserine mediated fusion between endothelial cell membrane and exosomes74. The process of angiogenesis also requires the activation of matrix metalloproteinases (MMPs) for the degradation of basement membrane, liberation of angiogenic factors, and sprouting of the capillaries. Tumor cells overexpress membrane bound molecule CD147, which is an extracellular MMPs inducer. Studies have shown that cancer cells (lung carcinoma, colon carcinoma, and pancreatic cancers) produce large amounts of CD147-positive MVs75,76 that can interact with the cells of TME and stimulate the production of MMPs. It is also observed that CD147-positive secretory vesicles of ovarian cancer cells promote angiogenic phenotype in endothelial cells (HUVECs) and pre-treatment of siRNA against CD147 suppressed their angiogenic potential77. Induction of endothelial cell (EC) migration and angiogenesis is mediated by the CCR1, CCL20, CXCL5, and MIF expression in ECs as evident from co-culturing them with ASTspan8 carrying exosomes78. Interestingly, it has also been found that D6.1A (tetraspanin Tspan8)-expressing tumor cells’ supernatant as well as D6.1A-containing exosomes, strongly induce angiogenesis79.
Additionally, it is also reported that hypoxic condition enhances the release of exosomes, which further promote microvascular endothelial cell migration and vasculogenesis. For example, hypoxia triggers the release of TF/VIIa bearing exosomes, which in turn increase the pro-angiogenic growth factor HB-EGF in ECs via ERK1/2-PAR2 dependent pathway80.
As an explanation of its capability to induce angiogenesis, exosomes has been found to modulate the many signaling pathways in a surprisingly novel mode. For instance, Notch signaling is an evolutionary conserved pathway that requires cell-to-cell contact for ligand-receptor interaction and further induction of cascade of its characteristics events. It has been shown that exosomes have signaling potential by transferring Dll-4 (ligand of notch receptor) to the neighbouring cells and incorporate it into the plasma membrane in vitro and in vivo. They have the ability to inhibit Notch signaling in vitro and appear to switch the endothelial cell phenotype toward tip cells phenotype, which in turn enhance vessel density29.
Interestingly, not every cell within the heterogenous tumor population secretes similar types of MVs, rather specific stem like cells; called cancer stem cells (CSCs) profoundly secrete specific MVs. For example, in the case of renal cancer, CD105 positive cells (CSCs) secrete MVs containing higher CD105. On co-culturing with HUVEC, only those MVs which are secreted by CSCs are able to form capillary like structures on matrigel and are able to enhance invasiveness of ECs. Furthermore, RNase pre-treatment of MVs reduced the above capabilities, which indicates that the content of these vesicles are mostly RNA molecules including miRNAs81. In this direction, it was further found that exogenous miR-9, previously reported to promote tumor cell motility and metastasis by repressing E-cadherin expression and increase VEGF transcription82, enhances EC migration and angiogenesis when exosomaly transferred to HUVECs. This exogenous miR-9 effectively reduced SOCS5 levels, leading to activation of JAK-STAT pathway83. Exosomes from colon cancer cells are able to induce mitosis after incorporation into endothelial cell (HUVECs) cytoplasm, which has been shown by immunostaining of both phospho-histone H3 (mitosis marker) and α-tubulin (mitotic spindle marker) in MV-treated HUVECs compared to control HUVECs84.
4.3 Role in Metastasis
Cancer metastasis involves the “leak”, or “spill” of potential cancer cells from the primary tumor, and settle down to other tissues in the body to establish a full blown secondary tumor85. It requires a cooperative interaction of tumor cells with non-tumor cells in the TME. Exosome mediated intercellular crosstalk plays a critical role in this coordinated process (Figure 1). Different tumor cells have distinct metastatic potential due to their genetic instability and exosomes are found to influence this genetic instability among tumor cells by transferring oncogenic sequences34. Exosomes are also found to enhance the metastatic potential of less metastatic melanoma cells B16-F1cells by transferring a metastasis marker (Met 72 tumor antigen) from highly metastatic B16 melanoma cells BL6-1086.
For successful evasion through extracellular matrix, tumor cells secrete MMPs or activators of MMPs like heat shock proteins (HSPs) for extracellular matrix remodelling. HSPs are also secreted through exosomes along with other proteins. It has been found that HSP90α together with annexin II in exosomes impart cell motility to their cancer cells via an interaction with an extracellular tissue plasminogen activator (tPA), which in turn activates protease plasmin87.Similar to extracellular matrix remodelling, vascular destabilization at the pre-metastatic niche is indispensable for cancer cell dissemination from the primary tumor site. Recently, exosomal miR-105 has been found to enhance tumor migration through enhancing vascular permeability by targeting tight junction protein ZO-1 (zonula occludens-1) in vascular endothelial cells88. The role of tumor derived exosomes in promoting metastatic potential of cancer cells is also supported by another elegant study in which, poorly metastasizing ASML-CD44vkd (CD44v-knockdown rat pancreatic adenocarcinoma BSp73ASML (ASMLwt) cells) cells regain metastatic capacity, when pre-treated with conditioned medium of ASMLwt cells containing exosomes. These exosomes target stromal and other cells of pre-metastatic organs to prepare metastatic niche to thrive tumor cells predominantly by transferring miRNAs (exosomal miR-494 and miR-542-3p which target cadherin-17)89. Furthermore Jung et al. also showed that conditioned media collected from ASMLwt, but not ASML-CD44vkd tumor cells promoted lymph node and lung metastasis of pancreatic cancer. Fractionation of conditioned media revealed that exosomes are the key factors which require CD44v for assembling soluble matrix90.
CAFs, an abundant stromal cell in the TME, support tumor growth by secreting several growth factors. They are found to stimulate breast cancer cells’ protrusive activity by exosomally transferring their endogenous Wnt11 to activate Wnt-planar cell polarity (PCP) in breast cancer cells91. These fibroblasts can be converted into myofibroblastic cells to support tumor growth, vascularization and metastasis. It has been shown that exosomal TGF-β was able to develop myofibroblastic phenotype in fibroblasts, through TGF-β-SMAD dependent signalling92. However, such differentiation was also found to occur in adipose derived mesenchymal stem cells via exosome mediated pathway in breast and ovarian cancer cells93,94. Recently, in an in vitro study, it has been demonstrated that 786‑0 renal cancer cell derived exosomes increased migration and invasion capacity of these cells by decreasing the adhesion ability and increasing the expression levels of CXCR4 and MMP‑995. Moreover, metastasis promoting epithelial-mesenchymal transition (EMT) related factors, such as vimentin, hepatoma-derived growth factor (HDGF) were found in the plasma membrane and annexin 2, CK2α, and moesin in the lumen of exosomes of bladder cancer respectively, suggesting their crucial involvement in metastatic process96.
4.4 Role in Drug Resistance
In the growing area of cancer research and treatment, development of chemoresistance is a decisive challenge for chemotherapy. Decreased drug uptake, increased drug efflux, activation of detoxifying systems, activation of DNA repair mechanisms and evasion of drug-induced apoptosis etc. are several defence mechanisms, which cancer cells can develop against the chemotherapy. However, the underlying molecular mechanism for chemoresistance still remains unclear. Thus, a more efficient strategy is required to target cancer cells, which are smarter than originally believed. Genetic and molecular studies have shown that most of the malignant cancer cells have amplified multi drug resistance (MDR) 1 and multidrug resistance associated protein (MRP) genes, which are the members of ATP-binding cassette transporter (ABC transporter) superfamily97. Now, a series of studies dictate that tumor derived exosomes added the layer of complexity in the complex nature of cancer by transmission of resistance from resistant cells to sensitive ones (Figure 1). Bebawy et al., showed the transmission of functional P-glycoprotein (P-gp) (MDR1) from drug resistant cancer cells (VLB100) to drug sensitive cancer cells (CCRF–CEM) over a co-culture period of 4 hours. They suggested that the expression of P-gp in recipient cell is not transcriptionally induced thus establishing a ‘non-genetic’ mechanism whereby MVs serve as a vector in the acquisition and spreading of MDR98. Recently, Wei-xian Chen et al. added another piece of evidence in transferring drug resistance via targeting MAPK pathway (putative target) through exosomal miRNAs of docetaxel resistant MCF-7 cells99. In a study with pulse chase and flow cytometric experimentation, vesicle shedding was represented as a potential mechanism of drug (doxorubicin) expulsion, which is found to be proportional to dose concentration100. Similarly, cisplatin (CDDP) was also found to expel out via exosomal pathway by CDDP-resistant cells, which possessed more putative CDDP transporters like ATP7A, ATP7B, and MRP2 (ABCB2) in order to escape from chemotherapeutic pressure. However, the routing of intercellular CDDP to exosomal pathway is not clear yet101. It has been shown that adriamycin-resistant MCF-7 cells’ derived MVs aided the resistance in recipient cells by transferring Ca2+ permeable channel TrpC5, which after incorporation induced the expression of P-gp102.
Exosome-antibody interaction is another way by which cancer cells evade chemotherapeutic pressure. It has been found that exosome antibody sequestration reduces the antibody-dependent cytotoxicity in cancer cells by immune effector cells103. Moreover, HER2-overexpressing breast cancer cell lines express a full-length HER2 molecule on exosomes, which can bind to the HER2 antibody Trastuzumab to nullify its effect on tumor proliferation104. Thus, it seems that exosomes, besides their permissive role in tumor growth, metastasis and angiogenesis, also program the tumor cells towards resistance to chemotherapy.
5. Conclusion and Future Direction
Exosomes are not just cellular debris but have a functional importance in cancer biology. In a pliant TME, where every cell communicates with each other through various ways, exosomes represent a new pathway to transport the information from donor to recipient cells. As an intercellular player, exosomes exhibit their ability to promote tumor growth, metastasis niche formation and provoking angiogenesis by carrying their specific cargo. Although, extensive researches on exosomes from the past few years have revealed their various new roles in cancer progression, a deep understanding of their biogenesis, sorting, secretion and uptake are still in its infancy. Moreover, few questions also remain obscured including “how recipient cells discriminate between the exosomes they need to take from a cohort of exosomes in a microenvironmental exosomal pool”. Thus, a better understanding is warranted for selective exosomal uptake by recipient cells, in order to exert a specific stimulation.
Exosomes comprise of bioactive components such as proteins, RNAs, miRNAs or lipids, and play a pivotal role in modulating tumor niche by controlling intercellular crosstalk.
Exosomes mediated intercellular communications and signals within TME regulate tumor growth, angiogenesis, metastasis and drug resistance.
Exosomal biogenesis, sorting, secretion and discriminative uptake from the microenvironmental exosomal pool by recipient cells are emerging areas and future directions of research.
Acknowledgments
This work was supported by different CSIR network projects and fellowships grants from CSIR, UGC, and MOES.
Footnotes
Conflict of interests: Authors have no potential conflict of interest.
Extracellular vesicles (EV); Tumor Microenvironment (TME); Carcinoma-associated fibroblasts (CAFs); Tumor-associated macrophages (TAM); Transforming growth factor-beta (TGF-b); Vascular endothelial growth factor (VEGF); Endothelial cell (EC); Matrix metalloproteinases (MMPs); Heat shock proteins (HSPs); Cancer stem cells (CSCs); Microvesicles (MVs); Epithelial-mesenchymal transition (EMT); Multi drug resistance-1 (MDR-1); Multidrug resistance associated protein (MRP); ATP-binding cassette transporter (ABC transporter)
DISCOVERIES is a peer-reviewed, open access, online, multidisciplinary and integrative journal, publishing high impact and innovative manuscripts from all areas related to MEDICINE, BIOLOGY and CHEMISTRY
References
- 1.Cell secretion and membrane fusion. Jena Bhanu P. Domestic animal endocrinology. 2005;29(1):145–65. doi: 10.1016/j.domaniend.2005.02.039. [DOI] [PubMed] [Google Scholar]
- 2.Proteomic profiling of exosomes: current perspectives. Simpson Richard J, Jensen Søren S, Lim Justin W E. Proteomics. 2008;8(19):4083–99. doi: 10.1002/pmic.200800109. [DOI] [PubMed] [Google Scholar]
- 3.Gateway to understanding microparticles: standardized isolation and identification of plasma membrane-derived vesicles. Dinkla Sip, Brock Roland, Joosten Irma, Bosman Giel J C G M. Nanomedicine (London, England) 2013;8(10):1657–68. doi: 10.2217/nnm.13.149. [DOI] [PubMed] [Google Scholar]
- 4.Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Pan B T, Johnstone R M. Cell. 1983;33(3):967–78. doi: 10.1016/0092-8674(83)90040-5. [DOI] [PubMed] [Google Scholar]
- 5.Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: demonstration of a pathway for receptor shedding. Harding C, Heuser J, Stahl P. European journal of cell biology. 1984;35(2):256–63. [PubMed] [Google Scholar]
- 6.Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). Johnstone R M, Adam M, Hammond J R, Orr L, Turbide C. The Journal of biological chemistry. 1987;262(19):9412–20. [PubMed] [Google Scholar]
- 7.Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Lee Yi, El Andaloussi Samir, Wood Matthew J A. Human molecular genetics. 2012;21(R1):R125–34. doi: 10.1093/hmg/dds317. [DOI] [PubMed] [Google Scholar]
- 8.Exosomes--vesicular carriers for intercellular communication. Simons Mikael, Raposo Graça. Current opinion in cell biology. 2009;21(4):575–81. doi: 10.1016/j.ceb.2009.03.007. [DOI] [PubMed] [Google Scholar]
- 9.Extracellular Microvesicles: The Need for Internationally Recognised Nomenclature and Stringent Purification Criteria. J. Simpson Richard, Mathivanan Suresh. Journal of Proteomics & Bioinformatics. 2012;05(02) [Google Scholar]
- 10.Quest for Cancer Biomarkers: Assaying Mutant Proteins and RNA that Provides the Much Needed Specificity. Mathivanan Suresh. Journal of Proteomics & Bioinformatics. 2012;05(11) [Google Scholar]
- 11.Exosomes: extracellular organelles important in intercellular communication. Mathivanan Suresh, Ji Hong, Simpson Richard J. Journal of proteomics. 2010;73(10):1907–20. doi: 10.1016/j.jprot.2010.06.006. [DOI] [PubMed] [Google Scholar]
- 12.Ectosomes released by human neutrophils are specialized functional units. Hess C, Sadallah S, Hefti A, Landmann R, Schifferli J A. Journal of immunology (Baltimore, Md. : 1950) 1999;163(8):4564–73. [PubMed] [Google Scholar]
- 13.Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Heijnen H F, Schiel A E, Fijnheer R, Geuze H J, Sixma J J. Blood. 1999;94(11):3791–9. [PubMed] [Google Scholar]
- 14.Exosomes: composition, biogenesis and function. Théry Clotilde, Zitvogel Laurence, Amigorena Sebastian. Nature reviews. Immunology. 2002;2(8):569–79. doi: 10.1038/nri855. [DOI] [PubMed] [Google Scholar]
- 15.Proteomic and biochemical analyses of human B cell-derived exosomes. Potential implications for their function and multivesicular body formation. Wubbolts Richard, Leckie Rachel S, Veenhuizen Peter T M, Schwarzmann Guenter, Möbius Wiebke, Hoernschemeyer Joerg, Slot Jan-Willem, Geuze Hans J, Stoorvogel Willem. The Journal of biological chemistry. 2003;278(13):10963–72. doi: 10.1074/jbc.M207550200. [DOI] [PubMed] [Google Scholar]
- 16.Exosome lipidomics unravels lipid sorting at the level of multivesicular bodies. Subra Caroline, Laulagnier Karine, Perret Bertrand, Record Michel. Biochimie. 2007;89(2):205–12. doi: 10.1016/j.biochi.2006.10.014. [DOI] [PubMed] [Google Scholar]
- 17.Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Trajkovic Katarina, Hsu Chieh, Chiantia Salvatore, Rajendran Lawrence, Wenzel Dirk, Wieland Felix, Schwille Petra, Brügger Britta, Simons Mikael. Science (New York, N.Y.) 2008;319(5867):1244–7. doi: 10.1126/science.1153124. [DOI] [PubMed] [Google Scholar]
- 18.ExoCarta 2012: database of exosomal proteins, RNA and lipids. Mathivanan Suresh, Fahner Cassie J, Reid Gavin E, Simpson Richard J. Nucleic acids research. 2012;40(Database issue):D1241–4. doi: 10.1093/nar/gkr828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Exosomal-like vesicles are present in human blood plasma. Caby Marie-Pierre, Lankar Danielle, Vincendeau-Scherrer Claude, Raposo Graça, Bonnerot Christian. International immunology. 2005;17(7):879–87. doi: 10.1093/intimm/dxh267. [DOI] [PubMed] [Google Scholar]
- 20.Identification and proteomic profiling of exosomes in human urine. Pisitkun Trairak, Shen Rong-Fong, Knepper Mark A. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(36):13368–73. doi: 10.1073/pnas.0403453101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Exosomes with immune modulatory features are present in human breast milk. Admyre Charlotte, Johansson Sara M, Qazi Khaleda Rahman, Filén Jan-Jonas, Lahesmaa Riitta, Norman Mikael, Neve Etienne P A, Scheynius Annika, Gabrielsson Susanne. Journal of immunology (Baltimore, Md. : 1950) 2007;179(3):1969–78. doi: 10.4049/jimmunol.179.3.1969. [DOI] [PubMed] [Google Scholar]
- 22.Pregnancy-Associated Exosomes and Their Modulation of T Cell Signaling. Taylor D. D., Akyol S., Gercel-Taylor C. The Journal of Immunology. 2006;176(3):1534-1542. doi: 10.4049/jimmunol.176.3.1534. [DOI] [PubMed] [Google Scholar]
- 23.Proteomic analysis of exosomes isolated from human malignant pleural effusions. Bard Martin P, Hegmans Joost P, Hemmes Annabrita, Luider Theo M, Willemsen Rob, Severijnen Lies-Anne A, van Meerbeeck Jan P, Burgers Sjaak A, Hoogsteden Henk C, Lambrecht Bart N. American journal of respiratory cell and molecular biology. 2004;31(1):114–21. doi: 10.1165/rcmb.2003-0238OC. [DOI] [PubMed] [Google Scholar]
- 24.Exosomes with major histocompatibility complex class II and co-stimulatory molecules are present in human BAL fluid. Admyre C., Grunewald J., Thyberg J., Gripenbäck S., Tornling G., Eklund A., Scheynius A., Gabrielsson S. European Respiratory Journal. 2003;22(4):578-583. doi: 10.1183/09031936.03.00041703. [DOI] [PubMed] [Google Scholar]
- 25.Association of citrullinated proteins with synovial exosomes. Skriner K, Adolph K, Jungblut P R, Burmester G R. Arthritis and rheumatism. 2006;54(12):3809–14. doi: 10.1002/art.22276. [DOI] [PubMed] [Google Scholar]
- 26.Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Théry Clotilde, Amigorena Sebastian, Raposo Graça, Clayton Aled. Current protocols in cell biology. 2006;Chapter 3:Unit 3.22. doi: 10.1002/0471143030.cb0322s30. [DOI] [PubMed] [Google Scholar]
- 27.Intercellular communication by exosome-derived microRNAs in cancer. Hannafon Bethany N, Ding Wei-Qun. International journal of molecular sciences. 2013;14(7):14240–69. doi: 10.3390/ijms140714240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Size distribution measurement of vesicles by atomic force microscopy. Kanno Takashi, Yamada Tadanori, Iwabuki Hidehiko, Tanaka Hiroyuki, Kuroda Shun'ichi, Tanizawa Katsuyuki, Kawai Tomoji. Analytical biochemistry. 2002;309(2):196–9. doi: 10.1016/s0003-2697(02)00291-9. [DOI] [PubMed] [Google Scholar]
- 29.New mechanism for Notch signaling to endothelium at a distance by Delta-like 4 incorporation into exosomes. Sheldon Helen, Heikamp Emily, Turley Helen, Dragovic Rebecca, Thomas Peter, Oon Chern Ein, Leek Russell, Edelmann Mariola, Kessler Benedikt, Sainson Richard C A, Sargent Ian, Li Ji-Liang, Harris Adrian L. Blood. 2010;116(13):2385–94. doi: 10.1182/blood-2009-08-239228. [DOI] [PubMed] [Google Scholar]
- 30.Extracellular vesicles: exosomes, microvesicles, and friends. Raposo Graça, Stoorvogel Willem. The Journal of cell biology. 2013;200(4):373–83. doi: 10.1083/jcb.201211138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Quantitative and qualitative flow cytometric analysis of nanosized cell-derived membrane vesicles. Nolte-'t Hoen Esther N M, van der Vlist Els J, Aalberts Marian, Mertens Hendrik C H, Bosch Berend Jan, Bartelink Willem, Mastrobattista Enrico, van Gaal Ethlinn V B, Stoorvogel Willem, Arkesteijn Ger J A, Wauben Marca H M. Nanomedicine : nanotechnology, biology, and medicine. 2012;8(5):712–20. doi: 10.1016/j.nano.2011.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qualitative analysis by high-resolution flow cytometry. van der Vlist Els J, Nolte-'t Hoen Esther N M, Stoorvogel Willem, Arkesteijn Ger J A, Wauben Marca H M. Nature Protocols. 2012;7(7):1311-1326. doi: 10.1038/nprot.2012.065. [DOI] [PubMed] [Google Scholar]
- 33.Exosomes as new vesicular lipid transporters involved in cell–cell communication and various pathophysiologies. Record Michel, Carayon Kevin, Poirot Marc, Silvente-Poirot Sandrine. Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 2014;1841(1):108-120. doi: 10.1016/j.bbalip.2013.10.004. [DOI] [PubMed] [Google Scholar]
- 34.Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Balaj Leonora, Lessard Ryan, Dai Lixin, Cho Yoon-Jae, Pomeroy Scott L., Breakefield Xandra O., Skog Johan. Nature Communications. 2011;2:180. doi: 10.1038/ncomms1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.The Tumor Suppressor PTEN Is Exported in Exosomes and Has Phosphatase Activity in Recipient Cells. Putz U., Howitt J., Doan A., Goh C.-P., Low L.-H., Silke J., Tan S.-S. Science Signaling. 2012;5(243):ra70-ra70. doi: 10.1126/scisignal.2003084. [DOI] [PubMed] [Google Scholar]
- 36.Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Valadi Hadi, Ekström Karin, Bossios Apostolos, Sjöstrand Margareta, Lee James J, Lötvall Jan O. Nature cell biology. 2007;9(6):654–9. doi: 10.1038/ncb1596. [DOI] [PubMed] [Google Scholar]
- 37.Transfer of the chemokine receptor CCR5 between cells by membrane-derived microparticles: a mechanism for cellular human immunodeficiency virus 1 infection. Mack M, Kleinschmidt A, Brühl H, Klier C, Nelson P J, Cihak J, Plachý J, Stangassinger M, Erfle V, Schlöndorff D. Nature medicine. 2000;6(7):769–75. doi: 10.1038/77498. [DOI] [PubMed] [Google Scholar]
- 38.Generation of novel, secreted epidermal growth factor receptor (EGFR/ErbB1) isoforms via metalloprotease-dependent ectodomain shedding and exosome secretion. Sanderson Michael P, Keller Sascha, Alonso Angel, Riedle Svenja, Dempsey Peter J, Altevogt Peter. Journal of cellular biochemistry. 2008;103(6):1783–97. doi: 10.1002/jcb.21569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Exosomes from human macrophages and dendritic cells contain enzymes for leukotriene biosynthesis and promote granulocyte migration. Esser Julia, Gehrmann Ulf, D'Alexandri Fabio Luiz, Hidalgo-Estévez Alicia M, Wheelock Craig E, Scheynius Annika, Gabrielsson Susanne, Rådmark Olof. The Journal of allergy and clinical immunology. 2010;126(5):1032–40, 1040.e1. doi: 10.1016/j.jaci.2010.06.039. [DOI] [PubMed] [Google Scholar]
- 40.Shedding of tissue factor (TF)-containing microparticles rather than alternatively spliced TF is the main source of TF activity released from human cancer cells. Yu J L, Rak J W. Journal of thrombosis and haemostasis : JTH. 2004;2(11):2065–7. doi: 10.1111/j.1538-7836.2004.00972.x. [DOI] [PubMed] [Google Scholar]
- 41.The origin, function, and diagnostic potential of RNA within extracellular vesicles present in human biological fluids. Taylor Douglas D, Gercel-Taylor Cicek. Frontiers in genetics. 2013;4:142. doi: 10.3389/fgene.2013.00142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Profiling of microRNAs in exosomes released from PC-3 prostate cancer cells. Hessvik Nina Pettersen, Phuyal Santosh, Brech Andreas, Sandvig Kirsten, Llorente Alicia. Biochimica et biophysica acta. 2012;1819(11-12):1154–63. doi: 10.1016/j.bbagrm.2012.08.016. [DOI] [PubMed] [Google Scholar]
- 43.Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. Ohshima Keiichi, Inoue Kanako, Fujiwara Akemi, Hatakeyama Keiichi, Kanto Kaori, Watanabe Yuko, Muramatsu Koji, Fukuda Yorikane, Ogura Shun-ichiro, Yamaguchi Ken, Mochizuki Tohru. PloS one. 2010;5(10):e13247. doi: 10.1371/journal.pone.0013247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hypoxic enhancement of exosome release by breast cancer cells. King Hamish W, Michael Michael Z, Gleadle Jonathan M. BMC cancer. 2012;12:421. doi: 10.1186/1471-2407-12-421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Glioma microvesicles carry selectively packaged coding and non-coding RNAs which alter gene expression in recipient cells. Li Cheryl C Y, Eaton Sally A, Young Paul E, Lee Maggie, Shuttleworth Rupert, Humphreys David T, Grau Georges E, Combes Valery, Bebawy Mary, Gong Joyce, Brammah Susan, Buckland Michael E, Suter Catherine M. RNA biology. 2013;10(8):1333–44. doi: 10.4161/rna.25281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ovarian cancer cell invasiveness is associated with discordant exosomal sequestration of Let-7 miRNA and miR-200. Kobayashi Miharu, Salomon Carlos, Tapia Jorge, Illanes Sebastian E, Mitchell Murray D, Rice Gregory E. Journal of translational medicine. 2014;12:4. doi: 10.1186/1479-5876-12-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Exosomes: a common pathway for a specialized function. van Niel Guillaume, Porto-Carreiro Isabel, Simoes Sabrina, Raposo Graça. Journal of biochemistry. 2006;140(1):13–21. doi: 10.1093/jb/mvj128. [DOI] [PubMed] [Google Scholar]
- 48.Hypoxia-inducible factors and RAB22A mediate formation of microvesicles that stimulate breast cancer invasion and metastasis. Wang Ting, Gilkes Daniele M, Takano Naoharu, Xiang Lisha, Luo Weibo, Bishop Corey J, Chaturvedi Pallavi, Green Jordan J, Semenza Gregg L. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(31):E3234–42. doi: 10.1073/pnas.1410041111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Rab27a and Rab27b control different steps of the exosome secretion pathway. Ostrowski Matias, Carmo Nuno B, Krumeich Sophie, Fanget Isabelle, Raposo Graça, Savina Ariel, Moita Catarina F, Schauer Kristine, Hume Alistair N, Freitas Rui P, Goud Bruno, Benaroch Philippe, Hacohen Nir, Fukuda Mitsunori, Desnos Claire, Seabra Miguel C, Darchen François, Amigorena Sebastian, Moita Luis F, Thery Clotilde. Nature cell biology. 2010;12(1):19–30; sup pp 1. doi: 10.1038/ncb2000. [DOI] [PubMed] [Google Scholar]
- 50.Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A–C. Hsu Chieh, Morohashi Yuichi, Yoshimura Shin-ichiro, Manrique-Hoyos Natalia, Jung SangYong, Lauterbach Marcel A., Bakhti Mostafa, Grønborg Mads, Möbius Wiebke, Rhee JeongSeop, Barr Francis A., Simons Mikael. The Journal of Cell Biology. 2010;189(2):223-232. doi: 10.1083/jcb.200911018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Active Wnt proteins are secreted on exosomes. Gross Julia Christina, Chaudhary Varun, Bartscherer Kerstin, Boutros Michael. Nature Cell Biology. 2012;14(10):1036-1045. doi: 10.1038/ncb2574. [DOI] [PubMed] [Google Scholar]
- 52.Regulation of exosome release by glycosphingolipids and flotillins. Phuyal Santosh, Hessvik Nina P., Skotland Tore, Sandvig Kirsten, Llorente Alicia. FEBS Journal. 2014;281(9):2214-2227. doi: 10.1111/febs.12775. [DOI] [PubMed] [Google Scholar]
- 53.Exosome Release Is Regulated by a Calcium-dependent Mechanism in K562 Cells. Savina Ariel, Furlán Marcelo, Vidal Michel, Colombo Maria I. Journal of Biological Chemistry. 2003;278(22):20083-20090. doi: 10.1074/jbc.M301642200. [DOI] [PubMed] [Google Scholar]
- 54.Anticancer drugs cause release of exosomes with heat shock proteins from human hepatocellular carcinoma cells that elicit effective natural killer cell antitumor responses in vitro. Lv Li-Hong, Wan Yun-Le, Lin Yan, Zhang Wei, Yang Mei, Li Guo-Lin, Lin Hao-Ming, Shang Chang-Zhen, Chen Ya-Jin, Min Jun. The Journal of biological chemistry. 2012;287(19):15874–85. doi: 10.1074/jbc.M112.340588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Thermal- and oxidative stress causes enhanced release of NKG2D ligand-bearing immunosuppressive exosomes in leukemia/lymphoma T and B cells. Hedlund Malin, Nagaeva Olga, Kargl Dominic, Baranov Vladimir, Mincheva-Nilsson Lucia. PloS one. 2011;6(2):e16899. doi: 10.1371/journal.pone.0016899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Regulation of exosome release from mammary epithelial and breast cancer cells - a new regulatory pathway. Riches Andrew, Campbell Elaine, Borger Eva, Powis Simon. European journal of cancer (Oxford, England : 1990) 2014;50(5):1025–34. doi: 10.1016/j.ejca.2013.12.019. [DOI] [PubMed] [Google Scholar]
- 57.Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Morelli Adrian E, Larregina Adriana T, Shufesky William J, Sullivan Mara L G, Stolz Donna Beer, Papworth Glenn D, Zahorchak Alan F, Logar Alison J, Wang Zhiliang, Watkins Simon C, Falo Louis D, Thomson Angus W. Blood. 2004;104(10):3257–66. doi: 10.1182/blood-2004-03-0824. [DOI] [PubMed] [Google Scholar]
- 58.Cellular internalization of exosomes occurs through phagocytosis. Feng Du, Zhao Wen-Long, Ye Yun-Ying, Bai Xiao-Chen, Liu Rui-Qin, Chang Lei-Fu, Zhou Qiang, Sui Sen-Fang. Traffic (Copenhagen, Denmark) 2010;11(5):675–87. doi: 10.1111/j.1600-0854.2010.01041.x. [DOI] [PubMed] [Google Scholar]
- 59.Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis. Fitzner Dirk, Schnaars Mareike, van Rossum Denise, Krishnamoorthy Gurumoorthy, Dibaj Payam, Bakhti Mostafa, Regen Tommy, Hanisch Uwe-Karsten, Simons Mikael. Journal of cell science. 2011;124(Pt 3):447–58. doi: 10.1242/jcs.074088. [DOI] [PubMed] [Google Scholar]
- 60.Microenvironmental pH is a key factor for exosome traffic in tumor cells. Parolini Isabella, Federici Cristina, Raggi Carla, Lugini Luana, Palleschi Simonetta, De Milito Angelo, Coscia Carolina, Iessi Elisabetta, Logozzi Mariantonia, Molinari Agnese, Colone Marisa, Tatti Massimo, Sargiacomo Massimo, Fais Stefano. The Journal of biological chemistry. 2009;284(49):34211–22. doi: 10.1074/jbc.M109.041152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Implication of tumor microenvironment in chemoresistance: tumor-associated stromal cells protect tumor cells from cell death. Castells Magali, Thibault Benoît, Delord Jean-Pierre, Couderc Bettina. International journal of molecular sciences. 2012;13(8):9545–71. doi: 10.3390/ijms13089545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Competitive cell interactions in cancer: a cellular tug of war. Wagstaff Laura, Kolahgar Golnar, Piddini Eugenia. Trends in cell biology. 2013;23(4):160–7. doi: 10.1016/j.tcb.2012.11.002. [DOI] [PubMed] [Google Scholar]
- 63.Competitive interactions of cancer cells and normal cells via secretory microRNAs. Kosaka Nobuyoshi, Iguchi Haruhisa, Yoshioka Yusuke, Hagiwara Keitaro, Takeshita Fumitaka, Ochiya Takahiro. The Journal of biological chemistry. 2012;287(2):1397–405. doi: 10.1074/jbc.M111.288662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Interactions between exosomes from breast cancer cells and primary mammary epithelial cells leads to generation of reactive oxygen species which induce DNA damage response, stabilization of p53 and autophagy in epithelial cells. Dutta Sujoy, Warshall Case, Bandyopadhyay Chirosree, Dutta Dipanjan, Chandran Bala. PloS one. 2014;9(5):e97580. doi: 10.1371/journal.pone.0097580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Critical physiological and pathological functions of Forkhead Box O tumor suppressors. Dumitrascu Georgiana R, Bucur Octavian. Discoveries. 2013:e5. doi: 10.15190/d.2013.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tumor-derived microvesicles induce, expand and up-regulate biological activities of human regulatory T cells (Treg). Szajnik Marta, Czystowska Malgorzata, Szczepanski Miroslaw J, Mandapathil Magis, Whiteside Theresa L. PloS one. 2010;5(7):e11469. doi: 10.1371/journal.pone.0011469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Surface-bound TGF-beta1 on effusion-derived exosomes participates in maintenance of number and suppressive function of regulatory T-cells in malignant effusions. Wada Junji, Onishi Hideya, Suzuki Hiroyuki, Yamasaki Akio, Nagai Shuntaro, Morisaki Takashi, Katano Mitsuo. Anticancer research. 2010;30(9):3747–57. [PubMed] [Google Scholar]
- 68.Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles. Andreola Giovanna, Rivoltini Licia, Castelli Chiara, Huber Veronica, Perego Paola, Deho Paola, Squarcina Paola, Accornero Paola, Lozupone Francesco, Lugini Luana, Stringaro Annarita, Molinari Agnese, Arancia Giuseppe, Gentile Massimo, Parmiani Giorgio, Fais Stefano. The Journal of experimental medicine. 2002;195(10):1303–16. doi: 10.1084/jem.20011624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Human colorectal cancer cells induce T-cell death through release of proapoptotic microvesicles: role in immune escape. Huber Veronica, Fais Stefano, Iero Manuela, Lugini Luana, Canese Paola, Squarcina Paola, Zaccheddu Annamaria, Colone Marisa, Arancia Giuseppe, Gentile Massimo, Seregni Ettore, Valenti Roberta, Ballabio Giuseppina, Belli Filiberto, Leo Ermanno, Parmiani Giorgio, Rivoltini Licia. Gastroenterology. 2005;128(7):1796–804. doi: 10.1053/j.gastro.2005.03.045. [DOI] [PubMed] [Google Scholar]
- 70.Cancer exosomes express CD39 and CD73, which suppress T cells through adenosine production. Clayton Aled, Al-Taei Saly, Webber Jason, Mason Malcolm D, Tabi Zsuzsanna. Journal of immunology (Baltimore, Md. : 1950) 2011;187(2):676–83. doi: 10.4049/jimmunol.1003884. [DOI] [PubMed] [Google Scholar]
- 71.Murine mammary carcinoma exosomes promote tumor growth by suppression of NK cell function. Liu Cunren, Yu Shaohua, Zinn Kurt, Wang Jianhua, Zhang Liming, Jia Yujiang, Kappes John C, Barnes Stephen, Kimberly Robert P, Grizzle William E, Zhang Huang-Ge. Journal of immunology (Baltimore, Md. : 1950) 2006;176(3):1375–85. doi: 10.4049/jimmunol.176.3.1375. [DOI] [PubMed] [Google Scholar]
- 72.Immune modulation of T-cell and NK (natural killer) cell activities by TEXs (tumour-derived exosomes). Whiteside Theresa L. Biochemical Society transactions. 2013;41(1):245–51. doi: 10.1042/BST20120265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Microvesicle-delivery miR-150 promotes tumorigenesis by up-regulating VEGF, and the neutralization of miR-150 attenuate tumor development. Liu Yuchen, Zhao Luming, Li Dameng, Yin Yuan, Zhang Chen-Yu, Li Jing, Zhang Yujing. Protein & cell. 2013;4(12):932–41. doi: 10.1007/s13238-013-3092-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Endothelial expression of autocrine VEGF upon the uptake of tumor-derived microvesicles containing oncogenic EGFR. Al-Nedawi Khalid, Meehan Brian, Kerbel Robert S, Allison Anthony C, Rak Janusz. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(10):3794–9. doi: 10.1073/pnas.0804543106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.The microvesicle as a vehicle for EMMPRIN in tumor-stromal interactions. Sidhu Sukhvinder S, Mengistab Aklilu T, Tauscher Andrew N, LaVail Jennifer, Basbaum Carol. Oncogene. 2004;23(4):956–63. doi: 10.1038/sj.onc.1207070. [DOI] [PubMed] [Google Scholar]
- 76.Tumour-derived microvesicles carry several surface determinants and mRNA of tumour cells and transfer some of these determinants to monocytes. Baj-Krzyworzeka Monika, Szatanek Rafał, Weglarczyk Kazimierz, Baran Jarosław, Urbanowicz Barbara, Brański Piotr, Ratajczak Mariusz Z, Zembala Marek. Cancer immunology, immunotherapy : CII. 2006;55(7):808–18. doi: 10.1007/s00262-005-0075-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Tumor vesicle-associated CD147 modulates the angiogenic capability of endothelial cells. Millimaggi Danilo, Mari Marianna, D'Ascenzo Sandra, Carosa Eleonora, Jannini Emmanuele Angelo, Zucker Stanley, Carta Gaspare, Pavan Antonio, Dolo Vincenza. Neoplasia (New York, N.Y.) 2007;9(4):349–57. doi: 10.1593/neo.07133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Nazarenko Irina, Rana Sanyukta, Baumann Alexandra, McAlear Jessica, Hellwig Andrea, Trendelenburg Michael, Lochnit Günter, Preissner Klaus T, Zöller Margot. Cancer research. 2010;70(4):1668–78. doi: 10.1158/0008-5472.CAN-09-2470. [DOI] [PubMed] [Google Scholar]
- 79.Systemic induction of the angiogenesis switch by the tetraspanin D6.1A/CO-029. Gesierich Sabine, Berezovskiy Igor, Ryschich Eduard, Zöller Margot. Cancer research. 2006;66(14):7083–94. doi: 10.1158/0008-5472.CAN-06-0391. [DOI] [PubMed] [Google Scholar]
- 80.Hypoxia triggers a proangiogenic pathway involving cancer cell microvesicles and PAR-2-mediated heparin-binding EGF signaling in endothelial cells. Svensson Katrin J, Kucharzewska Paulina, Christianson Helena C, Sköld Stefan, Löfstedt Tobias, Johansson Maria C, Mörgelin Matthias, Bengzon Johan, Ruf Wolfram, Belting Mattias. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(32):13147–52. doi: 10.1073/pnas.1104261108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Grange Cristina, Tapparo Marta, Collino Federica, Vitillo Loriana, Damasco Christian, Deregibus Maria Chiara, Tetta Ciro, Bussolati Benedetta, Camussi Giovanni. Cancer research. 2011;71(15):5346–56. doi: 10.1158/0008-5472.CAN-11-0241. [DOI] [PubMed] [Google Scholar]
- 82.miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Ma Li, Young Jennifer, Prabhala Harsha, Pan Elizabeth, Mestdagh Pieter, Muth Daniel, Teruya-Feldstein Julie, Reinhardt Ferenc, Onder Tamer T, Valastyan Scott, Westermann Frank, Speleman Frank, Vandesompele Jo, Weinberg Robert A. Nature cell biology. 2010;12(3):247–56. doi: 10.1038/ncb2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. Zhuang Guanglei, Wu Xiumin, Jiang Zhaoshi, Kasman Ian, Yao Jenny, Guan Yinghui, Oeh Jason, Modrusan Zora, Bais Carlos, Sampath Deepak, Ferrara Napoleone. The EMBO journal. 2012;31(17):3513–23. doi: 10.1038/emboj.2012.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Colorectal cancer cell-derived microvesicles are enriched in cell cycle-related mRNAs that promote proliferation of endothelial cells. Hong Bok Sil, Cho Ji-Hoon, Kim Hyunjung, Choi Eun-Jeong, Rho Sangchul, Kim Jongmin, Kim Ji Hyun, Choi Dong-Sic, Kim Yoon-Keun, Hwang Daehee, Gho Yong Song. BMC genomics. 2009;10:556. doi: 10.1186/1471-2164-10-556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cell motility and cytoskeletal regulation in invasion and metastasis. Kedrin Dmitriy, van Rheenen Jacco, Hernandez Lorena, Condeelis John, Segall Jeffrey E. Journal of mammary gland biology and neoplasia. 2007;12(2-3):143–52. doi: 10.1007/s10911-007-9046-4. [DOI] [PubMed] [Google Scholar]
- 86.Epigenetic transfer of metastatic activity by uptake of highly metastatic B16 melanoma cell-released exosomes. Hao S, Ye Z, Li F, Meng Q, Qureshi M, Yang J, Xiang J. Experimental oncology. 2006;28(2):126–31. [PubMed] [Google Scholar]
- 87.Secretion of extracellular hsp90alpha via exosomes increases cancer cell motility: a role for plasminogen activation. McCready Jessica, Sims Jessica D, Chan Doug, Jay Daniel G. BMC cancer. 2010;10:294. doi: 10.1186/1471-2407-10-294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Zhou Weiying, Fong Miranda Y, Min Yongfen, Somlo George, Liu Liang, Palomares Melanie R, Yu Yang, Chow Amy, O'Connor Sean Timothy Francis, Chin Andrew R, Yen Yun, Wang Yafan, Marcusson Eric G, Chu Peiguo, Wu Jun, Wu Xiwei, Li Arthur Xuejun, Li Zhuo, Gao Hanlin, Ren Xiubao, Boldin Mark P, Lin Pengnian Charles, Wang Shizhen Emily. Cancer cell. 2014;25(4):501–15. doi: 10.1016/j.ccr.2014.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Exosomal tumor microRNA modulates premetastatic organ cells. Rana Sanyukta, Malinowska Kamilla, Zöller Margot. Neoplasia (New York, N.Y.) 2013;15(3):281–95. doi: 10.1593/neo.122010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.CD44v6 dependence of premetastatic niche preparation by exosomes. Jung Thorsten, Castellana Donatello, Klingbeil Pamela, Cuesta Hernández Ines, Vitacolonna Mario, Orlicky David J, Roffler Steve R, Brodt Pnina, Zöller Margot. Neoplasia (New York, N.Y.) 2009;11(10):1093–105. doi: 10.1593/neo.09822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Luga Valbona, Zhang Liang, Viloria-Petit Alicia M, Ogunjimi Abiodun A, Inanlou Mohammad R, Chiu Elaine, Buchanan Marguerite, Hosein Abdel Nasser, Basik Mark, Wrana Jeffrey L. Cell. 2012;151(7):1542–56. doi: 10.1016/j.cell.2012.11.024. [DOI] [PubMed] [Google Scholar]
- 92.Cancer exosomes trigger fibroblast to myofibroblast differentiation. Webber Jason, Steadman Robert, Mason Malcolm D, Tabi Zsuzsanna, Clayton Aled. Cancer research. 2010;70(23):9621–30. doi: 10.1158/0008-5472.CAN-10-1722. [DOI] [PubMed] [Google Scholar]
- 93.Exosomes from breast cancer cells can convert adipose tissue-derived mesenchymal stem cells into myofibroblast-like cells. Cho Jung Ah, Park Ho, Lim Eun Hye, Lee Kyo Won. International journal of oncology. 2012;40(1):130–8. doi: 10.3892/ijo.2011.1193. [DOI] [PubMed] [Google Scholar]
- 94.Exosomes from ovarian cancer cells induce adipose tissue-derived mesenchymal stem cells to acquire the physical and functional characteristics of tumor-supporting myofibroblasts. Cho Jung Ah, Park Ho, Lim Eun Hye, Kim Kye Hyun, Choi Joong Sub, Lee Jung Hoon, Shin Jae Wook, Lee Kyo Won. Gynecologic oncology. 2011;123(2):379–86. doi: 10.1016/j.ygyno.2011.08.005. [DOI] [PubMed] [Google Scholar]
- 95.786-0 Renal cancer cell line-derived exosomes promote 786-0 cell migration and invasionin vitro. Chen Gang, Zhang Yao, Wu Xiaohou. Oncology letters. 2014;7(5):1576–1580. doi: 10.3892/ol.2014.1962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Quantitative proteomics of fractionated membrane and lumen exosome proteins from isogenic metastatic and nonmetastatic bladder cancer cells reveal differential expression of EMT factors. Jeppesen Dennis Kjølhede, Nawrocki Arkadiusz, Jensen Steffen Grann, Thorsen Kasper, Whitehead Bradley, Howard Kenneth A, Dyrskjøt Lars, Ørntoft Torben Falck, Larsen Martin R, Ostenfeld Marie Stampe. Proteomics. 2014;14(6):699–712. doi: 10.1002/pmic.201300452. [DOI] [PubMed] [Google Scholar]
- 97.Mechanisms of multidrug resistance in cancer. Gillet Jean-Pierre, Gottesman Michael M. Methods in molecular biology (Clifton, N.J.) 2010;596:47–76. doi: 10.1007/978-1-60761-416-6_4. [DOI] [PubMed] [Google Scholar]
- 98.Membrane microparticles mediate transfer of P-glycoprotein to drug sensitive cancer cells. Bebawy M, Combes V, Lee E, Jaiswal R, Gong J, Bonhoure A, Grau G E R. Leukemia. 2009;23(9):1643–9. doi: 10.1038/leu.2009.76. [DOI] [PubMed] [Google Scholar]
- 99.Exosomes from docetaxel-resistant breast cancer cells alter chemosensitivity by delivering microRNAs. Chen Wei-Xian, Cai Yan-Qin, Lv Meng-Meng, Chen Lin, Zhong Shan-Liang, Ma Teng-Fei, Zhao Jian-Hua, Tang Jin-Hai. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. 2014;35(10):9649–59. doi: 10.1007/s13277-014-2242-0. [DOI] [PubMed] [Google Scholar]
- 100.Expulsion of small molecules in vesicles shed by cancer cells: association with gene expression and chemosensitivity profiles. Shedden Kerby, Xie Xue Tao, Chandaroy Parthapratim, Chang Young Tae, Rosania Gustavo R. Cancer research. 2003;63(15):4331–7. [PubMed] [Google Scholar]
- 101.Abnormal lysosomal trafficking and enhanced exosomal export of cisplatin in drug-resistant human ovarian carcinoma cells. Safaei Roohangiz, Larson Barrett J, Cheng Timothy C, Gibson Michael A, Otani Shinji, Naerdemann Wiltrud, Howell Stephen B. Molecular cancer therapeutics. 2005;4(10):1595–604. doi: 10.1158/1535-7163.MCT-05-0102. [DOI] [PubMed] [Google Scholar]
- 102.Essential role for TrpC5-containing extracellular vesicles in breast cancer with chemotherapeutic resistance. Ma Xin, Chen Zhen, Hua Dong, He Dongxu, Wang Linjun, Zhang Peng, Wang Junqi, Cai Yanfei, Gao Caiji, Zhang Xiaodong, Zhang Fangfang, Wang Teng, Hong Tingting, Jin Linfang, Qi Xiaowei, Chen Shuxian, Gu Xiaoting, Yang Dangtong, Pan Qiongxi, Zhu Yifei, Chen Yun, Chen Daozhen, Jiang Liwen, Han Xiaofeng, Zhang Yanyun, Jin Jian, Yao Xiaoqiang. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(17):6389–94. doi: 10.1073/pnas.1400272111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Tumour exosomes inhibit binding of tumour-reactive antibodies to tumour cells and reduce ADCC. Battke Christina, Ruiss Romana, Welsch Ulrich, Wimberger Pauline, Lang Stephan, Jochum Simon, Zeidler Reinhard. Cancer immunology, immunotherapy : CII. 2011;60(5):639–48. doi: 10.1007/s00262-011-0979-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. Ciravolo Valentina, Huber Veronica, Ghedini Gaia C, Venturelli Elisabetta, Bianchi Francesca, Campiglio Manuela, Morelli Daniele, Villa Antonello, Della Mina Pamela, Menard Sylvie, Filipazzi Paola, Rivoltini Licia, Tagliabue Elda, Pupa Serenella M. Journal of cellular physiology. 2012;227(2):658–67. doi: 10.1002/jcp.22773. [DOI] [PubMed] [Google Scholar]

